US20230022157A1 - Pharmaceutical compounds for the treatment of complement factor d medical disorders - Google Patents

Pharmaceutical compounds for the treatment of complement factor d medical disorders Download PDF

Info

Publication number
US20230022157A1
US20230022157A1 US17/267,362 US201917267362A US2023022157A1 US 20230022157 A1 US20230022157 A1 US 20230022157A1 US 201917267362 A US201917267362 A US 201917267362A US 2023022157 A1 US2023022157 A1 US 2023022157A1
Authority
US
United States
Prior art keywords
alkyl
compound
formula
haloalkyl
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/267,362
Other languages
English (en)
Inventor
Jason Allan Wiles
Venkat Rao Gadhachanda
Kyle J. Eastman
Godwin Pais
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Achillion Pharmaceuticals Inc
Original Assignee
Achillion Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Achillion Pharmaceuticals Inc filed Critical Achillion Pharmaceuticals Inc
Priority to US17/267,362 priority Critical patent/US20230022157A1/en
Assigned to ACHILLION PHARMACEUTICALS, INC. reassignment ACHILLION PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PAIS, GODWIN, EASTMAN, KYLE J., GADHACHANDA, Venkat Rao, WILES, JASON ALLAN
Publication of US20230022157A1 publication Critical patent/US20230022157A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D451/00Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof
    • C07D451/02Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof containing not further condensed 8-azabicyclo [3.2.1] octane or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane; Cyclic acetals thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/052Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • This invention provides aryl, heteroaryl, and heterocyclic drugs to treat medical disorders, such as complement-mediated disorders, including Complement Factor D mediated disorders.
  • the complement system is a part of the innFate immune system which does not adapt to changes over the course of the host's life, but is recruited and used by the adaptive immune system. For example, it assists, or complements, the ability of antibodies and phagocytic cells to clear pathogens.
  • This sophisticated regulatory pathway allows rapid reaction to pathogenic organisms while protecting host cells from destruction.
  • Over thirty proteins and protein fragments make up the complement system. These proteins act through opsonization (enhancing phagocytosis of antigens), chemotaxis (attracting macrophages and neutrophils), cell lysis (rupturing membranes of foreign cells), and agglutination (clustering and binding of pathogens together).
  • Complement Factor D plays an early and central role in activation of the alternative pathway of the complement cascade. Activation of the alternative complement pathway is initiated by spontaneous hydrolysis of a thioester bond within the C3 protein to produce C3(H 2 O), which associates with Factor B to form the C3(H 2 O)B complex.
  • Complement Factor D acts to cleave Factor B within the C3(H 2 O)B complex to form Ba and Bb. The Bb fragment remains associated with C3(H 2 O) to form the alternative pathway C3 convertase C3(H 2 O)Bb.
  • C3b generated by any of the C3 convertases also associates with Factor B to form C3bB, which Factor D cleaves to generate the later stage alternative pathway C3 convertase C3bBb.
  • This latter form of the alternative pathway C3 convertase may provide important downstream amplification within all three of the defined complement pathways, leading ultimately to the recruitment and assembly of additional factors in the complement cascade pathway, including the cleavage of C5 to C5a and C5b.
  • C5b acts in the assembly of factors C6, C7, C8, and C9 into the membrane attack complex, which can destroy pathogenic cells by lysing the cell.
  • complement The dysfunction of or excessive activation of complement has been linked to certain autoimmune, inflammatory, and neurodegenerative diseases, as well as ischemia-reperfusion injury and cancer.
  • activation of the alternative pathway of the complement cascade contributes to the production of C3a and C5a, both potent anaphylatoxins, which also have roles in a number of inflammatory disorders. Therefore, in some instances, it is desirable to decrease the response of the complement pathway, including the alternative complement pathway.
  • disorders mediated by the complement pathway include age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), multiple sclerosis, and rheumatoid arthritis.
  • AMD age-related macular degeneration
  • PNH paroxysmal nocturnal hemoglobinuria
  • multiple sclerosis multiple sclerosis
  • rheumatoid arthritis Some examples include age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), multiple sclerosis, and
  • C3G is a recently defined entity comprised of dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) which encompasses a population of chronic kidney diseases wherein elevated activity of the alternative complement pathway and terminal complement pathway results in glomerular deposits made solely of complement C3 and no immunoglobulin (Ig).
  • DDD dense deposit disease
  • C3GN C3 glomerulonephritis
  • Immune-complex membranoproliferative glomerulonephritis is a renal disease which shares many clinical, pathologic, genetic and laboratory features with C3G, and therefore can be considered a sister disease of C3G.
  • an underlying disease or disorder most commonly infections, autoimmune diseases, or monoclonal gammopathies—are identified to which the renal disease is secondary.
  • Patients with idiopathic IC-MPGN can have low C3 and normal C4 levels, similar to those observed in C3G, as well as many of the same genetic or acquired factors that are associated with abnormal alternative pathway activity.
  • aHUS hemolytic uremic syndrome
  • HUS hemolytic uremic syndrome
  • NMO neuromyelitis optica
  • MG myasthenia gravis
  • MG myasthenia gravis
  • fatty liver nonalcoholic steatohepatitis
  • NASH nonalcoholic steatohepatitis
  • liver inflammation cirrhosis
  • liver failure dermatomyositis
  • amyotrophic lateral sclerosis amyotrophic lateral sclerosis
  • Factor D is an attractive target for inhibition or regulation of the complement cascade due to its early and essential role in the alternative complement pathway, and for its potential role in signal amplification within the classical and lectin complement pathways. Inhibition of Factor D effectively interrupts the pathway and attenuates the formation of the membrane attack complex.
  • BioCryst Pharmaceuticals Inc. U.S. Pat. No. 6,653,340 titled “Compounds useful in the complement, coagulate and kallikrein pathways and method for their preparation” describes fused bicyclic ring compounds that are inhibitors of Factor D.
  • US Patent Application 2019-0142802 assigned to BioCryst Pharmaceuticals describes benzopyrazole compounds for the treatment of aberrant complement disorders. Additional patents assigned to BioCryst for the treatment and prevention of complement disorders include granted U.S. Pat. No. 10,125,102 and US Applications US2018-0362458. Development of BioCryst's Factor D inhibitor BCX1470 was discontinued due to lack of specificity and short half-life of the compound.
  • Novartis PCT patent publication WO2012/093101 titled “Indole compounds or analogues thereof useful for the treatment of age-related macular degeneration” describes certain Factor D inhibitors. Additional Factor D inhibitors are described in Novartis PCT patent publications WO2012093101, WO2013/164802, WO2013/192345, WO2014/002051, WO2014/002052, WO2014/002053, WO2014/002054, WO2014/002057, WO2014/002058, WO2014/002059, WO2014/005150, WO2014/009833, WO2014/143638, WO2015/009616, WO2015/009977, WO2015/066241, and WO2016088082.
  • PCT patent publication WO2017/098328 titled “Therapeutic Inhibitory Compounds” describes various Factor D inhibitors with variations in the central core heterocyclic ring.
  • PCT patent publication WO2018/015818 is also titled “Therapeutic Inhibitory Compounds” and describes Factor D inhibitors without a cyclic central core.
  • Alexion Pharmaceuticals PCT patent publication WO1995/029697 titled “Methods and compositions for the treatment of glomerulonephritis and other inflammatory diseases” discloses antibodies directed to C5 of the complement pathway for the treatment of glomerulonephritis and inflammatory conditions involving pathologic activation of the complement system.
  • Alexion Pharmaceutical's anti-C5 antibody eculizumab Soliris® is currently the only complement-specific antibody on the market, and is the first and only approved treatment for paroxysmal nocturnal hemoglobinuria (PNH).
  • This invention includes a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition, wherein at least one substituent on the A group is an aryl, heteroaryl or heterocycle substituent.
  • the compound or its salt or composition, as described herein is used to treat a medical disorder which is an inflammatory or immune condition, a disorder mediated by the complement cascade (including a dysfunctional cascade), a disorder or abnormality of a cell that adversely affects the ability of the cell to engage in or respond to normal complement activity including the alternative complement pathway, or an undesired complement-mediated response to a medical treatment, such as surgery or other medical procedure or a pharmaceutical or biopharmaceutical drug administration, a blood transfusion, or other allogenic tissue or fluid administration.
  • a medical disorder which is an inflammatory or immune condition, a disorder mediated by the complement cascade (including a dysfunctional cascade), a disorder or abnormality of a cell that adversely affects the ability of the cell to engage in or respond to normal complement activity including the alternative complement pathway, or an undesired complement-mediated response to a medical treatment, such as surgery or other medical procedure or a pharmaceutical or biopharmaceutical drug administration, a blood transfusion, or other allogenic tissue or fluid administration.
  • a method for the treatment of such a disorder includes the administration of an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XII, or Formula XXIII or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition, as described in more detail below.
  • compounds are provided that have minimal effect on BSEP (bile salt export pump protein) (e.g., with an IC 50 of greater than about 20, 30, 40, 50, 60, 75 or 100 ⁇ M or greater), or with a therapeutic index of BSEP relative to complement D inhibition (e.g., IC 50 inhibition of BSEP/IC 50 inhibition of complement D inhibitor), of about at least 50, 100, 200, 300, 400, 500, 750 or 1000 or greater).
  • BSEP inhibition correlates with cholestatic drug-induced liver injury.
  • the compounds of the present invention exhibit reduced hydrolysis of the amide bond between the C ring and the B ring in vivo, for example, by including a proline that has a cis-substituent relative to the proline-carbonyl bond directed toward the B-ring.
  • the cis-substituent is in the R 3 position or the R 2 position or is a group that combines R 3 and R 2 .
  • a B-ring substituent in the position ortho to the amide may decrease the potential for formation of reactive metabolites.
  • an R 301 acylated embodiment of an active compound of the invention that exhibits extended half-life or other advantageous pharmacokinetic properties, which may be achieved by albumin stabilization in vivo.
  • the acylated analog can include several linking moieties in linear, branched or cyclic manner.
  • either one or a series of amino acids is used as a linker to a terminal fatty acid.
  • a non-natural amino acid such as one described below, for example 8-amino-3,6-dioxaoctanoic acid (one or several in sequence) is covalently bound to the selected complement D inhibitor of the present invention through a functional group such as a carboxylic acid, sulfonyl, hydroxyl or amino group.
  • a functional group such as a carboxylic acid, sulfonyl, hydroxyl or amino group.
  • the 8-amino-3,6-dioxaoctanoic acid or similar molecule is covalently linked to an aliphatic acid, including but not limited to a C 16 , C 18 , C 20 aliphatic acid, or a dicarboxylic acid, including but not limited to a C 8 , C 10 , C 12 , C 14 , C 16 , C 18 or C 20 diacid.
  • One or more amino acids can also be used in the selected configuration to add length or functionality.
  • nonlimiting embodiments include the use of a divalent linker moiety such as a dicarboxylic acid, amino acid, diamine, hydroxycarboxylic acid, hydroxyamine, di-hydroxy compound, or other compound that has at least two functional groups that can link the parent molecule with another linking moiety, and which may be albumin stabilized in vivo.
  • a divalent linker moiety such as a dicarboxylic acid, amino acid, diamine, hydroxycarboxylic acid, hydroxyamine, di-hydroxy compound, or other compound that has at least two functional groups that can link the parent molecule with another linking moiety, and which may be albumin stabilized in vivo.
  • 2, 3, 4 or 5 linking moieties are covalently bound in sequence, branched or cyclic fashion to the parent compound.
  • an R 301 acyl group is located in a position of the active compound that does not significantly adversely affect the complement D inhibition of the molecule, for example, as (i) a substituent on the R 32 group or (ii) a substituent on a C-ring, such as proline, or as a substituent on a substituent on the C-ring, such as on an R 1 , R 2 or R 3 substituent, including for example, on a bridged moiety such as a fused cyclopropyl on the proline ring.
  • the acyl group has an aliphatic or heteroaliphatic carbon range of C 12 , C 14 , C 16 , C 18 , C 20 , C 22 or C 24 .
  • the disorder is associated with the alternative complement cascade pathway. In yet another embodiment, the disorder is associated with the complement classical pathway. In a further embodiment, the disorder is associated with the complement lectin pathway. Alternatively, the active compound or its salt or prodrug may act through a different mechanism of action than the complement cascade to treat the disorder described herein.
  • a method is provided for treating a host, typically a human, with a disorder mediated by the complement system, that includes administration of a prophylactic antibiotic or vaccine to reduce the possibility of a bacterial infection during the treatment using one of the compounds described herein. In certain embodiments, the host, typically a human, is given a prophylactic vaccine prior to, during or after treatment with one of the compounds described herein.
  • the host typically a human, is given a prophylactic antibiotic prior to, during or after treatment with one of the compounds described herein.
  • the infection is a meningococcal infection (e.g., septicemia and/or meningitis), an Aspergillus infection, or an infection due to an encapsulated organism, for example, Streptococcus pneumoniae or Haemophilus influenza type b (Hib), especially in children.
  • the vaccine or antibiotic is administered to the patient after contracting an infection due to, or concommitent with inhibition of the complement system.
  • x 0, 1, or 2;
  • n 1, 2, 3, or 4;
  • q 0, 1, 2, or 3;
  • R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ are independently selected at each occurrence, as appropriate, and only where a stable compound results, from hydrogen, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , halogen, hydroxyl, and C 1 -C 6 alkyl;
  • R 1 and R 2 are taken together to form a 3- to 6-membered carbocyclic ring;
  • R 2 and R 3 are taken together to form a 3- to 6-membered carbocyclic ring;
  • A1 is selected from:
  • A2 is selected from:
  • A3 is selected from:
  • A4 is selected from:
  • A5 is selected from:
  • B1 is selected from C 1 -C 6 alkyl, C 0 -C 4 alkyl-aryl, C 0 -C 4 alkyl-heteroaryl, and C 0 -C 4 alkyl-heterocycle wherein each B1 can be optionally substituted with 1, 2, 3, or 4 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy;
  • B2 is selected from:
  • B2 is selected from:
  • R is hydrogen or C 1 -C 4 alkyl
  • R 5 is selected from C 1 -C 3 alkyl, —NR 9 R 10 , and —NR 9 OR 10 ;
  • R 5 is —C 1 -C 3 alkyl-OR 9 ;
  • each R 8 is independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 10 , —C 0 -C 4 alkylOR 9 , C 1 -C 6 haloalkyl, and C 1 -C 6 haloalkoxy;
  • R 8 is hydrogen
  • each R 9 and R 10 are independently selected from hydrogen and C 1 -C 4 alkyl
  • each R 11 is independently selected from C 1 -C 3 alkyl, —OR 9 , and —NR 9 R 10 ;
  • R 11 is hydrogen
  • each R 12 is independently selected from hydrogen, C 1 -C 3 alkyl, and —C(O)R 11 ;
  • each R 12 is independently hydrogen or —C(O)R 11 ;
  • R 13 is —NR 9 OR 10 ;
  • X 13 and X 14 are independently selected from N and CR 31 ;
  • R 22 is selected from —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , halogen, hydroxyl, and C 1 -C 6 alkyl;
  • R 23 is selected from hydrogen, C 1 -C 6 alkyl, —C 0 -C 4 alkyl-aryl, —C 0 -C 4 alkyl-heteroaryl, and —C 0 -C 4 alkyl-heterocycle;
  • each R 31 is independently selected from hydrogen, halogen, C 1 -C 6 alkyl, hydroxyl, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy;
  • R 32 is selected from aryl, heteroaryl, and heterocycle wherein the aryl, heteroaryl, or heterocycle ring can be optionally substituted with 1, 2, or 3 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —CO 2 R 11 , cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy;
  • R 33 is selected from
  • R 34 is selected from halogen, hydroxyl, C 1 -C 6 alkyl, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy.
  • compositions comprising a compound or salt of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, or Formula VIII together with a pharmaceutically acceptable carrier are also disclosed.
  • a compound of Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula X, Formula XXI, Formula XXII, or Formula XXIII is provided:
  • Q is CH or N
  • Q1 is CH 2 , O, NH, or NR 9 ;
  • w 0, 1, or 2;
  • A6 is selected from:
  • A7 is selected from:
  • A8 is selected from:
  • B3 is selected from:
  • B4 is selected from:
  • B5 is selected from:
  • R 35 is selected from
  • each R 36 is independently selected from C 1 -C 6 alkyl, halogen, and C 1 -C 6 haloalkyl;
  • R 37 is selected from C(O)R 11 , F, and CN;
  • R 38 is selected from
  • each R 39 is independently selected from hydrogen, C 1 -C 6 alkyl, halogen, and C 1 -C 6 haloalkyl;
  • R 40 is selected from
  • R 41 is selected from
  • each R 42 is independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 10 , —C 0 -C 4 alkylOR 9 , C 1 -C 6 haloalkyl, and C 1 -C 6 haloalkoxy; and
  • compositions comprising a compound or salt of Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII together with a pharmaceutically acceptable carrier are also disclosed.
  • the present invention thus includes at least the following features:
  • the compounds in any of the Formulas described herein include enantiomers, mixtures of enantiomers, diastereomers, tautomers, racemates and other isomers, such as rotamers, as if each is specifically described, unless otherwise indicated or otherwise excluded by context.
  • the present invention includes compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, or Formula XXI with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope, i.e., enriched.
  • isotopes examples include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 18 F, 31 P, 32 P, 35 S, 36 Cl, and 125 I respectively.
  • isotopically labelled compounds can be used in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F labeled compound may be particularly desirable for PET or SPECT studies.
  • Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • isotopes of hydrogen for example, deuterium (2H) and tritium (H) may optionally be used anywhere in described structures that achieves the desired result.
  • isotopes of carbon e.g., 13 C and 14 C, may be used.
  • the isotopic substitution is replacing hydrogen with a deuterium at one or more locations on the molecule to improve the performance of the drug, for example, the pharmacodynamics, pharmacokinetics, biodistribution, half-life, stability, AUC, Tmax, Cmax, etc.
  • the deuterium can be bound to carbon in a location of bond breakage during metabolism (an ⁇ -deuterium kinetic isotope effect) or next to or near the site of bond breakage (a ⁇ -deuterium kinetic isotope effect).
  • Isotopic substitutions for example deuterium substitutions, can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted with deuterium.
  • the isotope is 80, 85, 90, 95 or 99% or more enriched in an isotope at any location of interest.
  • deuterium is 80, 85, 90, 95 or 99% enriched at a desired location. Unless otherwise stated, the enrichment at any point is above natural abundance, and in an embodiment is enough to alter a detectable property of the drug in a human.
  • the substitution of a hydrogen atom for a deuterium atom can be provided in any of A1, A2, A3, A4, A5, B1, and B2. In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within any R group. In one embodiment the R group is selected from any of R, R 1 , R 1′ , R 2 , R 2′ , R 3 , R 3′ , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 22 , R 23 , R 31 , R 32 , R 33 , R 34 , R 201 , and R 301 .
  • the alkyl residue may be deuterated (in nonlimiting embodiments, CDH 2 , CD 2 H, CD 3 , CD 2 CD 3 , CHDCH 2 D, CH 2 CD 3 , CHDCHD 2 , OCDH 2 , OCD 2 H, or OCD 3 etc.).
  • an R group has a “′” or an “a” designation, which in one embodiment can be deuterated.
  • the unsubstituted methylene carbon may be deuterated.
  • the substitution of a hydrogen atom for a deuterium atom can be provided in any of A1, A2, A3, A4, A5, A6, A7, A8, B1, B2, B3, B4, or B5. In one embodiment, the substitution of a hydrogen atom for a deuterium atom occurs within any R group.
  • the R group is selected from any of R, R 1 , R 1′ , R 2 , R 2′ , R 3 , R 3′ , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 22 , R 23 , R 31 , R 32 , R 33 , R 34 , R 35 , R 36 , R 38 , R 39 , R 40 , R 41 , R 42 , R 201 , and R 301 .
  • the alkyl residue may be deuterated (in nonlimiting embodiments, CDH 2 , CD 2 H, CD 3 , CD 2 CD 3 , CHDCH 2 D, CH 2 CD 3 , CHDCHD 2 , OCDH 2 , OCD 2 H, or OCD 3 etc.).
  • an R group has a “′” or an “a” designation, which in one embodiment can be deuterated.
  • the unsubstituted methylene carbon may be deuterated.
  • the compound of the present invention may form a solvate with solvents (including water). Therefore, in one embodiment, the invention includes a solvated form of the active compound.
  • solvate refers to a molecular complex of a compound of the present invention (including a salt thereof) with one or more solvent molecules. Nonlimiting examples of solvents are water, ethanol, dimethyl sulfoxide, acetone and other common organic solvents.
  • hydrate refers to a molecular complex comprising a compound of the invention and water.
  • Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 O, d 6 -acetone, d 6 -DMSO.
  • a solvate can be in a liquid or solid form.
  • a dash (“-”) that is not between two letters or symbols is used to indicate a point of attachment for a substituent.
  • —(C ⁇ O)NH 2 is attached through carbon of the keto (C ⁇ O) group.
  • substituted means that any one or more hydrogens on the designated atom or group is replaced with a moiety selected from the indicated group, provided that the designated atom's normal valence is not exceeded and the resulting compound is stable.
  • substituent is oxo (i.e., ⁇ O) then two hydrogens on the atom are replaced.
  • oxo i.e., ⁇ O
  • a pyridyl group substituted by oxo is a pyridone.
  • a stable active compound refers to a compound that can be isolated and can be formulated into a dosage form with a shelf life of at least one month.
  • a stable manufacturing intermediate or precursor to an active compound is stable if it does not degrade within the period needed for reaction or other use.
  • a stable moiety or substituent group is one that does not degrade, react or fall apart within the period necessary for use.
  • Nonlimiting examples of unstable moieties are those that combine heteroatoms in an unstable arrangement, as typically known and identifiable to those of skill in the art.
  • Any suitable group may be present on a “substituted” or “optionally substituted” position that forms a stable molecule and meets the desired purpose of the invention and includes, but is not limited to, e.g., halogen (which can independently be F, Cl, Br or I); cyano; hydroxyl; nitro; azido; alkanoyl (such as a C 2 -C 6 alkanoyl group); carboxamide; alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy such as phenoxy; thioalkyl including those having one or more thioether linkages; alkylsulfinyl; alkylsulfonyl groups including those having one or more sulfonyl linkages; aryl (e.g., phenyl, biphenyl, naphthyl, or the like, each ring either substituted or unsubstituted); arylalkyl having for
  • “optionally substituted” includes one or more substituents independently selected from halogen, hydroxyl, amino, cyano, —CHO, —COOH, —CONH 2 , alkyl including C 1 -C 6 alkyl, alkenyl including C 2 -C 6 alkenyl, alkynyl including C 2 -C 6 alkynyl, —C 1 -C 6 alkoxy, alkanoyl including C 2 -C 6 alkanoyl, (mono- and di-C 1 -C 6 alkylamino)C 0 -C 2 alkyl, haloalkyl including C 1 -C 6 haloalkyl, hydoxyC 1 -C 6 alkyl, ester, carbamate, urea, sulfonamide, —C 1 -C 6 alkyl(heterocyclo), C 1 -C 6 alkyl
  • Alkyl is a branched, straight chain, or cyclic saturated aliphatic hydrocarbon group. In one embodiment, the alkyl contains from 1 to about 12 carbon atoms, more generally from 1 to about 6 carbon atoms or from 1 to about 4 carbon atoms. In one embodiment, the alkyl contains from 1 to about 8 carbon atoms. In certain embodiments, the alkyl is C 1 -C 2 , C 1 -C 3 , C 1 -C 4 , C 1 -C 5 or C 1 -C 6 .
  • the specified ranges as used herein indicate an alkyl group having each member of the range described as an independent species.
  • C 1 -C 6 alkyl indicates a straight or branched alkyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species.
  • C 1 -C 4 alkyl indicates a straight or branched alkyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species.
  • C 0 -C n alkyl is used herein in conjunction with another group, for example, (C 3 -C 7 cycloalkyl)C 0 -C 4 alkyl, or —C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl), the indicated group, in this case cycloalkyl, is either directly bound by a single covalent bond (C 0 alkyl), or attached by an alkyl chain in this case 1, 2, 3, or 4 carbon atoms.
  • Alkyls can also be attached via other groups such as heteroatoms as in —O—C 0 -C 4 alkyl(C 3 -C 7 cycloalkyl).
  • alkyl examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, tert-pentyl, neopentyl, n-hexyl, 2-methylpentane, 3-methylpentane, 2,2-dimethylbutane, 2,3-dimethylbutane, and hexyl.
  • “Aliphatic” refers to a saturated or unsaturated, straight, branched, or cyclic hydrocarbon. “Aliphatic” is intended herein to include, but is not limited to, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties, and thus incorporates each of these definitions.
  • “aliphatic” is used to indicate those aliphatic groups having 1-20 carbon atoms. The aliphatic chain can be, for example, mono-unsaturated, di-unsaturated, tri-unsaturated, polyunsaturated, or alkynyl.
  • Unsaturated aliphatic groups can be in a cis or trans configuration.
  • the aliphatic group contains from 1 to about 12 carbon atoms, more generally from 1 to about 6 carbon atoms or from 1 to about 4 carbon atoms.
  • the aliphatic group contains from 1 to about 8 carbon atoms.
  • the aliphatic group is C 1 -C 2 , C 1 -C 3 , C 1 -C 4 , C 1 -C 5 or C 1 -C 6 .
  • the specified ranges as used herein indicate an aliphatic group having each member of the range described as an independent species.
  • C 1 -C 6 aliphatic as used herein indicates a straight or branched alkyl, alkenyl, or alkynyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species.
  • C 1 -C 4 aliphatic as used herein indicates a straight or branched alkyl, alkenyl, or alkynyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species.
  • the aliphatic group is substituted with one or more functional groups that results in the formation of a stable moiety.
  • heteroaliphatic refers to an aliphatic moiety that contains at least one heteroatom in the chain, for example, an amine, carbonyl, carboxy, oxo, thio, phosphate, phosphonate, nitrogen, phosphorus, silicon, or boron atoms in place of a carbon atom.
  • the only heteroatom is nitrogen.
  • the only heteroatom is oxygen.
  • the only heteroatom is sulfur.
  • Heteroaliphatic is intended herein to include, but is not limited to, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycle, and heterocycloalkynyl moieties.
  • heteroaliphatic is used to indicate a heteroaliphatic group (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms.
  • the heteroaliphatic group is optionally substituted in a manner that results in the formation of a stable moiety.
  • Nonlimiting examples of heteroaliphatic moieties are polyethylene glycol, polyalkylene glycol, amide, polyamide, polylactide, polyglycolide, thioether, ether, alkyl-heterocycle-alkyl, —O-alkyl-O-alkyl, alkyl-O-haloalkyl, etc.
  • alk When a term is used that includes “alk” it should be understood that “cycloalkyl” or “carbocyclic” can be considered part of the definition, unless unambiguously excluded by the context.
  • alkyl, alkenyl, alkynyl, alkoxy, alkanoyl, alkenloxy, haloalkyl, etc. can all be considered to include the cyclic forms of alkyl, unless unambiguously excluded by context.
  • Alkenyl is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon double bonds that may occur at a stable point along the chain. Nonlimiting examples are C 2 -C 6 alkenyl, C 2 -C 7 alkenyl, C 2 -C 6 alkenyl, C 2 -C 5 alkenyl and C 2 -C 4 alkenyl.
  • the specified ranges as used herein indicate an alkenyl group having each member of the range described as an independent species, as described above for the alkyl moiety. Examples of alkenyl include, but are not limited to, ethenyl and propenyl.
  • Alkynyl is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon triple bonds that may occur at any stable point along the chain, for example, C 2 -C 6 alkynyl or C 2 -C 6 alkynyl.
  • the specified ranges as used herein indicate an alkynyl group having each member of the range described as an independent species, as described above for the alkyl moiety.
  • alkynyl examples include, but are not limited to, ethynyl, propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl and 5-hexynyl.
  • Alkoxy is an alkyl group as defined above covalently bound through an oxygen bridge (—O—).
  • alkoxy include, but are not limited to, methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, 2-butoxy, t-butoxy, n-pentoxy, 2-pentoxy, 3-pentoxy, isopentoxy, neopentoxy, n-hexoxy, 2-hexoxy, 3-hexoxy, and 3-methylpentoxy.
  • an “alkylthio” or a “thioalkyl” group is an alkyl group as defined above with the indicated number of carbon atoms covalently bound through a sulfur bridge (—S—). In one embodiment, the alkoxy group is optionally substituted as described above.
  • Alkenyloxy is an alkenyl group as defined covalently bound to the group it substitutes by an oxygen bridge (—O—).
  • Alkanoyl is an alkyl group as defined above covalently bound through a carbonyl (C ⁇ O) bridge.
  • the carbonyl carbon is included in the number of carbons, that is C 2 alkanoyl is a CH 3 (C ⁇ O)— group.
  • the alkanoyl group is optionally substituted as described
  • Haloalkyl indicates both branched and straight-chain alkyl groups substituted with 1 or more halogen atoms, up to the maximum allowable number of halogen atoms.
  • Examples of haloalkyl include, but are not limited to, trifluoromethyl, monofluoromethyl, difluoromethyl, 2-fluoroethyl, and penta-fluoroethyl.
  • Haloalkoxy indicates a haloalkyl group as defined herein attached through an oxygen bridge (oxygen of an alcohol radical).
  • Halo or “halogen” indicates independently, any of fluoro, chloro, bromo or iodo.
  • Aryl indicates an aromatic group containing only carbon in the aromatic ring or rings.
  • the aryl group contains 1 to 3 separate or fused rings and is 6 to 14 or 18 ring atoms, without heteroatoms as ring members.
  • such aryl groups may be further substituted with carbon or non-carbon atoms or groups. Such substitution may include fusion to a 4 to 7 or a 5 to 7-membered saturated or partially unsaturated cyclic group that optionally contains 1, 2 or 3 heteroatoms independently selected from N, O, B, P, Si and S, to form, for example, a 3,4-methylenedioxyphenyl group.
  • Aryl groups include, for example, phenyl and naphthyl, including 1-naphthyl and 2-naphthyl. In one embodiment, aryl groups are pendant.
  • An example of a pendant ring is a phenyl group substituted with a phenyl group.
  • heterocycle refers to saturated and partially saturated heteroatom-containing ring radicals, where the heteroatoms may be selected from N, S, and O.
  • heterocycle includes monocyclic 3-12 membered rings, as well as bicyclic 5-16 membered ring systems (which can include fused, bridged, or spiro, bicyclic ring systems). It does not include rings containing —O—O—. —O—S—, or —S—S— portions.
  • saturated heterocycle groups include saturated 4- to 7-membered monocyclic groups containing 1 to 4 nitrogen atoms [e.g.
  • pyrrolidinyl imidazolidinyl, piperidinyl, pyrrolinyl, azetidinyl, piperazinyl, and pyrazolidinyl]; saturated 4 to 6-membered monocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl]; saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms [e.g., thiazolidinyl].
  • partially saturated heterocycle radicals include but are not limited to, dihydrothienyl, dihydropyranyl, dihydrofuryl, and dihydrothiazolyl.
  • Examples of partially saturated and saturated heterocycle groups include but are not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, pyrazolidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, thiazolidinyl, dihydrothienyl, 2,3-dihydro-benzo[1,4]dioxanyl, indolinyl, isoindolinyl, dihydrobenzothienyl, dihydrobenzofuryl, isochromanyl, chromanyl, 1,2-dihydroquinolyl, 1,2,3,4-tetrahydro-isoquinolyl, 1,2,3,4-tetrahydro-quinolyl, 2,3,4,4a,9,9a-hexahydro-1H-3-aza-fluorenyl, 5,6,7-trihydro-1,2,4-triazolo[3,4-a]isoquinolyl
  • “Bicyclic heterocycle” includes groups wherein the heterocyclic radical is fused with an aryl radical wherein the point of attachment is the heterocycle ring. “Bicyclic heterocycle” also includes heterocyclic radicals that are fused with a carbocycle radical. For example partially unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, for example, indoline, isoindoline, partially unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, partially unsaturated condensed heterocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, and saturated condensed heterocyclic group containing 1 to 2 oxygen or sulfur atoms.
  • bicyclic heterocycles include:
  • bicyclic heterocycle includes cis and trans diastereomers.
  • Non-limiting examples of chiral bicyclic heterocycles include:
  • Heteroaryl refers to a stable monocyclic, bicyclic, or multicyclic aromatic ring which contains from 1 to 3, or in some embodiments from 1, 2, or 3 heteroatoms selected from N, O, S, B, and P (and typically selected from N, O, and S) with remaining ring atoms being carbon, or a stable bicyclic or tricyclic system containing at least one 5, 6, or 7 membered aromatic ring which contains from 1 to 3, or in some embodiments from 1 to 2, heteroatoms selected from N, O, S, B or P with remaining ring atoms being carbon.
  • the only heteroatom is nitrogen.
  • the only heteroatom is oxygen.
  • the only heteroatom is sulfur.
  • Monocyclic heteroaryl groups typically have from 5 or 6 ring atoms.
  • bicyclic heteroaryl groups are 8- to 10-membered heteroaryl groups, that is, groups containing 8 or 10 ring atoms in which one 5, 6, or 7 member aromatic ring is fused to a second aromatic or non-aromatic ring wherein the point of attachment is the aromatic ring.
  • the total number of S and O atoms in the heteroaryl group exceeds 1, these heteroatoms are not adjacent to one another.
  • the total number of S and O atoms in the heteroaryl group is not more than 2.
  • the total number of S and O atoms in the aromatic heterocycle is not more than 1.
  • heteroaryl groups include, but are not limited to, pyridinyl (including, for example, 2-hydroxypyridinyl), imidazolyl, imidazopyridinyl, pyrimidinyl (including, for example, 4-hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, furyl, thienyl, isoxazolyl, thiazolyl, oxadiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazoly
  • Heterocycloalkyl is a fully saturated heterocycle as defined herein. It may have, for example, include 1, 2, 3, or 4 heteroatoms independently selected from N, S, O, Si and B with the remaining ring atoms being carbon. In a typical embodiment, nitrogen is the heteroatom. Monocyclic heterocycloalkyl groups typically have from 3 to about 8 ring atoms or from 4 to 6 ring atoms.
  • a “dosage form” means a unit of administration of an active agent.
  • dosage forms include tablets, capsules, injections, suspensions, liquids, emulsions, implants, particles, spheres, creams, ointments, suppositories, inhalable forms, transdermal forms, buccal, sublingual, topical, gel, mucosal, and the like.
  • a “dosage form” can also include an implant, for example an optical implant.
  • “Pharmaceutical compositions” are compositions comprising at least one active agent, and at least one other substance, such as a carrier. “Pharmaceutical combinations” are combinations of at least two active agents which may be combined in a single dosage form or provided together in separate dosage forms with instructions that the active agents are to be used together to treat any disorder described herein.
  • a “pharmaceutically acceptable salt” is a derivative of the disclosed compound in which the parent compound is modified by making inorganic and organic, pharmaceutically acceptable, acid or base addition salts thereof.
  • the salts of the present compounds can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • salts of the present compounds further include solvates of the compounds and of the compound salts.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include salts which are acceptable for human consumption and the quaternary ammonium salts of the parent compound formed, for example, from inorganic or organic acids.
  • salts examples include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC—(CH 2 ) 1-4 —COOH, and the like, or using a different acid that produces the same counterion.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfa
  • carrier applied to pharmaceutical compositions/combinations of the invention refers to a diluent, excipient, or vehicle with which an active compound is provided.
  • a “pharmaceutically acceptable excipient” means an excipient that is useful in preparing a pharmaceutical composition/combination that is generally safe, acceptable for human consumption, and neither biologically nor otherwise inappropriate for administration to a host, typically a human. In one embodiment, an excipient is used that is acceptable for veterinary use.
  • a “patient” or “host” or “subject” is a human or non-human animal in need of treatment or prevention of any of the disorders as specifically described herein, including but not limited to by modulation of the complement Factor D pathway or with a condition that is treatable with one of the compounds described herein.
  • the host is a human.
  • a “patient” or “host” or “subject” also refers to for example, a mammal, primate (e.g., human), cows, sheep, goat, horse, dog, cat, rabbit, rat, mice, bird and the like.
  • a “prodrug” as used herein means a compound which when administered to a host in vivo is converted into a parent drug.
  • the term “parent drug” means any of the presently described chemical compounds herein.
  • Prodrugs can be used to achieve any desired effect, including to enhance properties of the parent drug or to improve the pharmaceutic or pharmacokinetic properties of the parent, including to increase the half-life of the drug in vivo.
  • Prodrug strategies provide choices in modulating the conditions for in vivo generation of the parent drug.
  • prodrug strategies include covalent attachment of removable groups, or removable portions of groups, for example, but not limited to acylation, phosphorylation, phosphonylation, phosphoramidate derivatives, amidation, reduction, oxidation, esterification, alkylation, other carboxy derivatives, sulfoxy or sulfone derivatives, carbonylation or anhydride, among others.
  • the prodrug renders the parent compound more lipophilic.
  • a prodrug can be provided that has several prodrug moieties in linear, branched or cyclic manner.
  • nonlimiting embodiments include the use of a divalent linker moiety such as a dicarboxylic acid, amino acid, diamine, hydroxycarboxylic acid, hydroxyamine, di-hydroxy compound, or other compound that has at least two functional groups that can link the parent molecule with another prodrug moiety, and is typically biodegradable in vivo.
  • a divalent linker moiety such as a dicarboxylic acid, amino acid, diamine, hydroxycarboxylic acid, hydroxyamine, di-hydroxy compound, or other compound that has at least two functional groups that can link the parent molecule with another prodrug moiety, and is typically biodegradable in vivo.
  • 2, 3, 4, or 5 prodrug biodegradable moieties are covalently bound in sequence, branched or cyclic fashion to the parent compound.
  • prodrugs according to the present invention are formed with:
  • a prodrug is provided by attaching a natural or non-natural amino acid to an appropriate functional moiety on the parent compound, for example, oxygen, nitrogen, or sulfur, and typically oxygen or nitrogen, usually in a manner such that the amino acid can be cleaved in vivo to provide the parent drug.
  • the amino acid can be used alone or covalently linked (straight, branched or cyclic) to one or more other prodrug moieties to modify the parent drug to achieve the desired performance, such as increased half-life, lipophilicity, or other drug delivery or pharmacokinetic properties.
  • the amino acid can be any compound with an amino group and a carboxylic acid, which includes an aliphatic amino acid, alkyl amino acid, aromatic amino acid, heteroaliphatic amino acid, heteroalkyl amino acid, or heterocyclic amino acid or heteroaryl amino acid.
  • “Providing a compound with at least one additional active agent,” for example, in one embodiment can mean that the compound and the additional active agent(s) are provided simultaneously in a single dosage form, provided concomitantly in separate dosage forms, or provided in separate dosage forms for administration.
  • the compound administrations are separated by some amount of time that is within the time in which both the compound and the at least one additional active agent are within the blood stream of a patient.
  • the compound and the additional active agent need not be prescribed for a patient by the same medical care worker.
  • the additional active agent or agents need not require a prescription.
  • Administration of the compound or the at least one additional active agent can occur via any appropriate route, for example, oral tablets, oral capsules, oral liquids, inhalation, injection, suppositories, parenteral, sublingual, buccal, intravenous, intraaortal, transdermal, polymeric controlled delivery, non-polymeric controlled delivery, nano or microparticles, liposomes, and/or topical contact.
  • the instructions for administration in a form of combination therapy is provided in the drug labeling.
  • a “therapeutically effective amount” of a pharmaceutical composition/combination of this invention means an amount effective, when administered to a host, provides a therapeutic benefit such as an amelioration of symptoms or reduction or dimunition of the disease itself.
  • a therapeutically effective amount is an amount sufficient to prevent a significant increase or will significantly reduce the detectable level of complement Factor D in the patient's blood, serum, or tissues.
  • any of the active compounds can be provided in its N-oxide form to a patient in need thereof.
  • an N-oxide of an active compound or a precursor of the active compound is used in a manufacturing scheme.
  • the N-oxide is a metabolite of administration of one of the active compounds herein, and may have independent activity.
  • the N-oxide can be formed by treating the compound of interest with an oxidizing agent, for example a suitable peroxyacid or peroxide, to generate an N-oxide compound.
  • a heteroaryl group for example a pyridyl group
  • an oxidizing agent such as sodium percarbonate
  • a rhenium-based catalyst under mild reaction conditions to generate an N-oxide compound.
  • oxidizing agent such as sodium percarbonate
  • protecting groups may be necessary to carry out the chemistry. See, Jain, S. L. et al., “Rhenium-Catalyzed Highly Efficient Oxidations of Tertiary Nitrogen Compounds to N-Oxides Using Sodium Percarbonate as Oxygen Source, Synlett, 2261-2663, 2006.
  • the N-oxide is in the A-Ring. In one embodiment the N-oxide is in the B-Ring. In one embodiment the N-oxide is on the R 32 group.
  • any of the active compounds with a sulfur can be provided in its sulfoxide or sulfone form to a patient in need thereof.
  • a sulfoxide or sulfone of one of the active compounds or a precursor of the active compound is used in a manufacturing scheme.
  • a sulfur atom in a selected compound as described herein can be oxidized to form a sulfoxide
  • TAPC 1,3,5-triazo-2,4,6-triphosphorine-2,2,4,4,6,6-tetrachloride
  • TAPC 1,3,5-triazo-2,4,6-triphosphorine-2,2,4,4,6,6-tetrachloride
  • Oxidation of sulfides with 30% hydrogen peroxide catalyzed by tantalum carbide provides sulfoxides in high yields, see, Kirihara, A., et al., “Tantalum Carbide or Niobium Carbide Catalyzed Oxidation of Sulfides with Hydrogen Peroxide: Highly Efficient and Chemoselective Syntheses of Sulfoxides and Sulfones”, Synlett, 1557-1561 (2010).
  • Sulfides can be oxidized to sulfones using, for example, niobium carbide as the catalyst, see, Kirihara, A., et al., “Tantalum Cardide or Niobium Carbide Catalyzed Oxidation of Sulfides with Hydrogen Peroxide: Highly Efficient and Chemoselective Syntheses of Sulfoxides and Sulfones”, Synlett, 1557-1561 (2010).
  • Urea-hydrogen peroxide adduct is a stable inexpensive and easily handled reagent for the oxidation of sulfides to sulfones, see Varma, R. S. and Naicker, K.
  • alkyl is a C 1 -C 10 alkyl, C 1 -C 9 alkyl, C 1 -C 6 alkyl, C 1 -C 7 alkyl, C 1 -C 6 alkyl, C 1 -C 5 alkyl, C 1 -C 4 alkyl, C 1 -C 3 alkyl, or C 1 -C 2 alkyl.
  • alkyl has one carbon
  • alkyl has two carbons.
  • alkyl has three carbons.
  • alkyl has four carbons.
  • alkyl has five carbons.
  • alkyl has six carbons.
  • alkyl include: methyl, ethyl, propyl, butyl, pentyl, and hexyl.
  • alkyl examples include: isopropyl, isobutyl, isopentyl, and isohexyl.
  • alkyl examples include: sec-butyl, sec-pentyl, and sec-hexyl.
  • alkyl examples include: tert-butyl, tert-pentyl, and tert-hexyl.
  • alkyl include: neopentyl, 3-pentyl, and active pentyl.
  • the “alkyl” group is optionally substituted.
  • haloalkyl is a C 1 -C 10 haloalkyl, C 1 -C 9 haloalkyl, C 1 -C 8 haloalkyl, C 1 -C 7 haloalkyl, C 1 -C 6 haloalkyl, C 1 -C 5 haloalkyl, C 1 -C 4 haloalkyl, C 1 -C 3 haloalkyl, and C 1 -C 2 haloalkyl.
  • haloalkyl has one carbon
  • haloalkyl has one carbon and one halogen.
  • haloalkyl has one carbon and two halogens.
  • haloalkyl has one carbon and three halogens.
  • haloalkyl has two carbons.
  • haloalkyl has three carbons.
  • haloalkyl has four carbons.
  • haloalkyl has five carbons.
  • haloalkyl has six carbons.
  • haloalkyl include:
  • haloalkyl include:
  • haloalkyl include:
  • haloalkyl include:
  • aryl is a 6 carbon aromatic group (phenyl)
  • aryl is a 10 carbon aromatic group (napthyl)
  • aryl is “substituted aryl”.
  • the “aryl” group is optionally substituted.
  • heteroaryl is a 5 membered aromatic group containing 1, 2, or 3, nitrogen atoms.
  • Non-limiting examples of 5 membered “heteroaryl” groups include pyrrole, furan, thiophene, pyrazole, imidazole, triazole, isoxazole, oxazole, oxadiazole, oxatriazole, isothiazole, thiazole, thiadiazole, and thiatriazole.
  • heteroaryl is a 6 membered aromatic group containing 1, 2, or 3 nitrogen atoms (i.e. pyridinyl, pyridazinyl, triazinyl, pyrimidinyl, and pyrazinyl).
  • Non-limiting examples of 6 membered “heteroaryl” groups with 1 or 2 nitrogen atoms include:
  • heteroaryl is a 9 membered bicyclic aromatic group containing 1 or 2 atoms selected from nitrogen, oxygen, and sulfur.
  • heteroaryl groups that are bicyclic include indole, benzofuran, isoindole, indazole, benzimidazole, azaindole, azaindazole, purine, isobenzofuran, benzothiophene, benzoisoxazole, benzoisothiazole, benzooxazole, and benzothiazole.
  • heteroaryl groups that are bicyclic include:
  • heteroaryl groups that are bicyclic include:
  • heteroaryl groups that are bicyclic include:
  • heteroaryl is a 10 membered bicyclic aromatic group containing 1 or 2 atoms selected from nitrogen, oxygen, and sulfur.
  • heteroaryl groups that are bicyclic include quinoline, isoquinoline, quinoxaline, phthalazine, quinazoline, cinnoline, and naphthyridine.
  • heteroaryl groups that are bicyclic include:
  • R 32 is selected from
  • R 32 is
  • halogen optionally substituted with 1, 2, or 3 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —CO 2 R 11 , cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy.
  • heteroaryl is tetrazole.
  • heteroaryl group is optionally substituted.
  • cycloalkyl is a C 3 -C 8 cycloalkyl, C 3 -C 7 cycloalkyl, C 3 -C 6 cycloalkyl, C 3 -C 5 cycloalkyl, C 3 -C 4 cycloalkyl, C 4 -C 8 cycloalkyl, C 5 -C 8 cycloalkyl, or C 6 -C 8 cycloalkyl.
  • cycloalkyl has three carbons.
  • cycloalkyl has four carbons.
  • cycloalkyl has five carbons.
  • cycloalkyl has six carbons.
  • cycloalkyl has seven carbons.
  • cycloalkyl has eight carbons.
  • cycloalkyl has nine carbons.
  • cycloalkyl has ten carbons.
  • cycloalkyl include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and cyclodecyl.
  • cycloalkyl group is optionally substituted.
  • heterocycle refers to a cyclic ring with one nitrogen and 3, 4, 5, 6, 7, or 8 carbon atoms.
  • heterocycle refers to a cyclic ring with one nitrogen and one oxygen and 3, 4, 5, 6, 7, or 8 carbon atoms.
  • heterocycle refers to a cyclic ring with two nitrogens and 3, 4, 5, 6, 7, or 8 carbon atoms.
  • heterocycle refers to a cyclic ring with one oxygen and 3, 4, 5, 6, 7, or 8 carbon atoms.
  • heterocycle refers to a cyclic ring with one sulfur and 3, 4, 5, 6, 7, or 8 carbon atoms.
  • heterocycle examples include aziridine, oxirane, thiirane, azetidine, 1,3-diazetidine, oxetane, and thietane.
  • heterocycle examples include pyrrolidine, 3-pyrroline, 2-pyrroline, pyrazolidine, and imidazolidine.
  • heterocycle examples include tetrahydrofuran, 1,3-dioxolane, tetrahydrothiophene, 1,2-oxathiolane, and 1,3-oxathiolane.
  • heterocycle examples include piperidine, piperazine, tetrahydropyran, 1,4-dioxane, thiane, 1,3-dithiane, 1,4-dithiane, morpholine, and thiomorpholine.
  • heterocycle also include:
  • heterocycle includes:
  • heterocycle includes:
  • heterocycle also include:
  • heterocycle also include:
  • heterocycle includes:
  • heterocycle includes:
  • heterocycle group is optionally substituted.
  • x 0, 1, or 2;
  • n 1, 2, 3, or 4;
  • q 0, 1, 2, or 3;
  • R 1 , R 1′ , R 2 , R 2′ , R 3 , and R 3′ are independently selected at each occurrence, as appropriate, and only where a stable compound results, from hydrogen, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , halogen, hydroxyl, and C 1 -C 6 alkyl;
  • R 1 and R 2 are taken together to form a 3- to 6-membered carbocyclic ring;
  • R 2 and R 3 are taken together to form a 3- to 6-membered carbocyclic ring;
  • A1 is selected from:
  • A2 is selected from:
  • A3 is selected from:
  • A4 is selected from:
  • A5 is selected from:
  • B1 is C 0 -C 4 alkyl-heteroaryl wherein each B1 can be optionally substituted with 1, 2, 3, or 4 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy;
  • B2 is selected from:
  • R is hydrogen or C 1 -C 4 alkyl
  • R 5 is selected from C 1 -C 3 alkyl, —NR 9 R 10 , —NR 9 OR 10 , and —C 1 -C 3 alkyl-OR 9 ;
  • each R 8 is independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 10 , —C 0 -C 4 alkylOR 9 , C 1 -C 6 haloalkyl, and C 1 -C 6 haloalkoxy;
  • each R 9 and R 10 are independently selected from hydrogen and C 1 -C 4 alkyl
  • each R 11 is independently selected from C 1 -C 3 alkyl, —OR 9 , and —NR 9 R 10 ;
  • R 11 is hydrogen
  • each R 12 is independently selected from hydrogen, C 1 -C 3 alkyl, and —C(O)R 11 ; in certain embodiments each R 12 is independently hydrogen or —C(O)R 11 ;
  • R 13 is —NR 9 OR 10 ;
  • X 13 and X 14 are independently selected from N and CR 31 ;
  • R 22 is selected from —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , halogen, hydroxyl, and C 1 -C 6 alkyl;
  • R 23 is selected from hydrogen, C 1 -C 6 alkyl, —C 0 -C 4 alkyl-aryl, —C 0 -C 4 alkyl-heteroaryl, and —C 0 -C 4 alkyl-heterocycle;
  • R 23 is selected from hydrogen, C 1 -C 6 alkyl, —C 0 -C 4 alkyl-aryl, —C 0 -C 4 alkyl-heteroaryl, C 2 -C 6 alkyl-OR 9 , and —C 0 -C 4 alkyl-heterocycle;
  • each R 31 is independently selected from hydrogen, halogen, C 1 -C 6 alkyl, hydroxyl, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy;
  • R 32 is selected from aryl, heteroaryl, and heterocycle wherein the aryl, heteroaryl, or heterocycle ring can be optionally substituted with 1, 2, or 3 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —CO 2 R 11 , cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy;
  • R 33 is selected from
  • R 34 is selected from halogen, hydroxyl, C 1 -C 6 alkyl, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy.
  • each R 8 is independently selected from hydrogen, halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, —C 0 -C 4 alkylNR 9 R 10 , —C 0 -C 4 alkylOR 9 , C 1 -C 6 haloalkyl, and C 1 -C 6 haloalkoxy.
  • each R 8 is independently selected from tetrazole, halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, —C 0 -C 4 alkylNR 9 R 10 , —C 0 -C 4 alkylOR 9 , C 1 -C 6 haloalkyl, and C 1 -C 6 haloalkoxy.
  • R 11 is hydrogen or C 1 -C 3 alkyl.
  • R 11 is hydrogen
  • each R 31 is independently selected from hydrogen, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl halogen, C 1 -C 6 alkyl, hydroxyl, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy;
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is B2, B3, B4, or B5.
  • B1 is selected from:
  • B1 is selected from:
  • B2 is selected from
  • B2 is selected from:
  • B3 is selected from:
  • B3 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B3 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B3 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B3 is selected from
  • B3 is selected from
  • B4 is selected from:
  • B5 is selected from:
  • B5 is selected from:
  • B5 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B5 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R is
  • A1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • A1 is selected from A2, A3, A4, A5, A6, A7 and A8.
  • A1 is selected from:
  • A2 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • A4 is selected from:
  • A5 is selected from:
  • A6 is selected from:
  • A7 is selected from:
  • A8 is selected from:
  • X 13 is CH and X 14 is selected from C(C 2 -C 6 alkenyl), C(C 2 -C 6 alkynyl), and C(C 1 -C 6 alkyl).
  • X 13 is N and X 14 is selected from C(C 2 -C 6 alkenyl), C(C 2 -C 6 alkynyl), and C(C 1 -C 6 alkyl).
  • X 14 is selected from C(C 2 -C 6 alkenyl), C(C 2 -C 6 alkynyl), and C(C 1 -C 6 alkyl).
  • X 14 is C(C 2 -C 6 alkenyl).
  • X 14 is C(C 2 -C 6 alkyl).
  • X 14 is C(CF 3 ).
  • X 14 is CH.
  • R 32 is
  • R 33 is
  • R 32 is selected from
  • R 32 is selected from R 33 , R 15 , and R 38 .
  • R 32 is selected from
  • R 33 is selected from
  • R 35 is selected from
  • R 38 is selected from
  • R 40 is selected from
  • R 41 is selected from
  • R 33 is selected from
  • each R 12 is independently selected from hydrogen and —C(O)R 11 .
  • R 33 is selected from
  • each R 12 is independently selected from hydrogen, C 1 -C 3 alkyl, and —C(O)R 11 .
  • a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula X, Formula XXI, Formula XXII, or Formula XXIII is provided wherein a single C—H bond is replaced with a R 201 group, wherein:
  • R 201 is selected from C 0 -C 3 alkyl-NR 9 R 15 , C 0 -C 3 alkyl-OR 15 , C 0 -C 3 alkyl-SR 15 ; C 0 -C 3 alkyl-heterocycle, -aliphatic-OR 15 , -aliphatic-SR 15 , and -aliphatic-NR 9 R 15 ; and wherein R 201 can be optionally substituted with R 301 , which can be directly linked to R 201 or can be linked to R 201 through an amino, hydroxyl, thio, carboxylic acid, phosphate, phosphonate, or sulfonate linkage; wherein the R 201 group is optionally substituted with 1, 2, or 3 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —
  • Each R 15 is independently selected from hydrogen, C 1 -C 3 alkyl, and —C(O)R 11 .
  • R 201 is selected from —(CH 2 ) 1-3 —O-heterocycle, —(CH 2 ) 1-3 —NH-heterocycle, or —(CH 2 ) 1-3 —NR 9 -heterocycle.
  • R 201 is selected from —(CH 2 ) 1-3 —NR 9 R 10 , —(CH 2 ) 1-3 —OR 9 , or —(CH 2 ) 1-3 -heterocycle.
  • R 201 is selected from —CH 2 —O-heterocycle, —CH 2 —NH-heterocycle, or —CH 2 —NR 9 -heterocycle.
  • R 201 is selected from —CH 2 —NR 9 R 10 , —CH 2 —OR 9 , or —CH 2 -heterocycle.
  • R 201 is selected from —(CH 2 ) 1-3 —NH 2 , —(CH 2 ) 1-3 —OH, or —(CH 2 ) 1-3 —OC 1 -C 6 alkyl.
  • R 201 is selected from —NR 9 R 10 , —OR 9 , or —NR 9 -heterocycle.
  • R 201 is selected from —O-heterocycle, —NH-heterocycle, or —NR 9 -heterocycle.
  • (CH 2 ) 1-3 is —CH 2 —
  • (CH 2 ) 1-3 is —CH 2 CH 2 —.
  • (CH 2 ) 1-3 is —CH 2 CH 2 CH 2 —.
  • n 1 or 2.
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • R 201 is selected from:
  • the compound of the present invention is selected from:
  • the compound of the present invention is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • the compound is selected from:
  • a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula X, Formula XXI, Formula XXII, or Formula XXIII is provided wherein one N—H or O—H bond is replaced with an R 301 group; wherein R 301 is selected from the following:
  • R 301 is selected from:
  • R 301 is selected from:
  • R 301 is
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are independently selected from: bond, polyethylene glycol, a natural amino acid, an unnatural amino acid,
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected from: bond, polyethylene glycol, a natural amino acid, an unnatural amino acid,
  • R 309 is selected from: alkyl, hydrogen,
  • n2 is independently selected at each instance from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20;
  • X 300 is selected from bond, —NH—, —N(alkyl)-, O, —CH 2 —O—, —CH 2 —NH—, and —CH 2 —N(alkyl)-.
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected to be bond.
  • none of R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected to be bond.
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected to be bond.
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected to be bond.
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected to be bond.
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected to be bond.
  • R 302 , R 303 , R 304 , R 305 , R 306 , R 307 , and R 308 are selected to be bond.
  • R 102 is C 1 -C 4 alkyl, fluorine, chlorine, or bromine.
  • R 1 is selected from F, Cl, Br, and C 1 -C 6 alkyl.
  • R 1 is selected from hydroxyl and C 1 -C 6 alkoxy.
  • R 1 is selected from C 2 -C 6 alkynyl, C 2 -C 6 alkanoyl, and C 1 -C 6 thioalkyl.
  • R 2 is hydrogen
  • R 2′ is selected from F, Cl, Br, and C 1 -C 6 alkyl.
  • R 2′ is selected from hydroxyl and C 1 -C 6 alkoxy.
  • R 2′ is selected from C 1 -C 6 alkyl.
  • R 2′ is selected from C 2 -C 6 alkynyl, C 2 -C 6 alkanoyl, and C 1 -C 6 thioalkyl.
  • R 1 is selected from aminoC 1 -C 6 alkyl and —C 0 -C 4 alkylNR 9 R 10 .
  • Non-limiting examples of A1 include:
  • A1 Additional non-limiting examples of A1 include:
  • A1 Additional non-limiting examples of A1 include:
  • A1 is selected from:
  • A1 is selected from:
  • A1 is selected from:
  • A1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • R 25 , R 26 , R 27 , and R 28 are independently selected from hydrogen, halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —COOH, cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy.
  • R 26 , R 27 , R 28 , and R 29 are independently selected from hydrogen, halogen, C 1 -C 3 alkyl, and C 1 -C 3 haloalkyl.
  • B1 moieties include, but are not limited to
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is selected from:
  • B1 is -alkyl-Si(alkyl) 3 or -alkyl-SF 5 .
  • B1 is a B ring substituted with oxo.
  • the B ring is a nitrogen containing heteroaryl group then the nitrogen may also be substituted as defined herein. For example:
  • B1 is selected from:
  • R 32 is a heteroaryl ring substituted with oxo as allowed by valence.
  • R 32 ring is a nitrogen containing heteroaryl group then the nitrogen may also be substituted as defined herein.
  • the nitrogen may also be substituted as defined herein.
  • R 32 is selected from:
  • R 32 is selected from aryl, heteroaryl, and heterocycle wherein the aryl, heteroaryl, or heterocycle ring can be optionally substituted with 1, 2, or 3 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —CO 2 R 11 , cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy, wherein the heterocycle ring is attached to the A ring through a carbon atom.
  • R 32 is selected from aryl, heteroaryl, and heterocycle wherein the aryl, heteroaryl, or heterocycle ring can be optionally substituted with 1, 2, or 3 groups independently selected from halogen, C 1 -C 3 alkyl, C 1 -C 3 haloalkyl, —C(O)R 11 , cyano, C 2 -C 6 alkanoyl, C 1 -C 6 alkoxy, —C 0 -C 4 alkylNR 9 R 12 , —C 0 -C 4 alkylOR 12 , C 1 -C 6 haloalkyl, —SO 2 R 11 , and C 1 -C 6 haloalkoxy, wherein the heterocycle ring is attached to the A ring through a carbon atom.
  • Active compounds described herein can be administered to a host in need thereof as the neat chemical, but are more typically administered as a pharmaceutical composition that includes an effective amount for a host, typically a human, in need of such treatment of an active compound as described herein or its pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof.
  • the disclosure provides pharmaceutical compositions comprising an effective amount of compound or pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof together with at least one pharmaceutically acceptable carrier for any of the uses described herein.
  • the pharmaceutical composition may contain a compound or salt as the only active agent, or, in an alternative embodiment, the compound and at least one additional active agent.
  • an effective amount of an active compound as described herein, or the active compound described herein in combination or alternation with, or preceded by, concomitant with or followed by another active agent can be used in an amount sufficient to (a) inhibit the progression of a disorder mediated by the complement pathway, including an inflammatory, immune, including an autoimmune, disorder or complement Factor D related disorder; (b) cause a regression of an inflammatory, immune, including an autoimmune, disorder or complement Factor D related disorder; (c) cause a cure of an inflammatory, immune, including an autoimmune, disorder or complement Factor D related disorder; or inhibit or prevent the development of an inflammatory, immune, including an autoimmune, disorder or complement Factor D related disorder.
  • an effective amount of an active compound or its salt or composition described herein will provide a sufficient amount of the active agent when administered to a patient to provide a clinical benefit.
  • the exact amount of the active compound or pharmaceutical composition described herein to be delivered to the host, typically a human, in need thereof, will be determined by the health care provider to achieve the desired clinical benefit.
  • the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of the active compound and optionally from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of an additional active agent in a unit dosage form.
  • Examples are dosage forms with at least about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 900, 1000, 1100, 1200, 1250, 1300, 1400, 1500, or 1600 mg of active compound, or its salt, N-oxide, isotopic analog, or prodrug.
  • the dosage form has at least about 1 mg, 5 mg, 10 mg, 25 mg, 50 mg, 75 mg, 100 mg, 200 mg, 400 mg, 500 mg, 600 mg, 1000 mg, 1200 mg, or 1600 mg of active compound, N-oxide, isotopic analog, prodrug, or its salt.
  • the amount of active compound in the dosage form is calculated without reference to the salt.
  • the dosage form can be administered, for example, once a day (q.d.), twice a day (b.i.d.), three times a day (t.i.d.), four times a day (q.i.d.), once every other day (Q2d), once every third day (Q3d), as needed, or any dosage schedule that provides treatment of a disorder described herein.
  • Compounds disclosed herein or used as described herein may be administered orally, topically, parenterally, by inhalation or spray, sublingually, via implant, including ocular implant, transdermally, via buccal administration, rectally, as an ophthalmic solution, injection, including ocular injection, intravenous, intra-aortal, intracranial, subdermal, intraperitoneal, subcutaneous, transnasal, sublingual, intrathecal, or rectal or by other means, in dosage unit formulations containing conventional pharmaceutically acceptable carriers.
  • the compound can be administered, as desired, for example, as a solution, suspension, or other formulation via intravitreal, intrastromal, intracameral, sub-tenon, sub-retinal, retro-bulbar, peribulbar, suprachorodial, subchorodial, chorodial, conjunctival, subconjunctival, episcleral, periocular, transscleral, retrobulbar, posterior juxtascleral, circumcorneal, or tear duct injections, or through a mucus, mucin, or a mucosal barrier, in an immediate or controlled release fashion or via an ocular device, injection, or topically administered formulation, for example a solution or suspension provided as an eye drop.
  • the pharmaceutical composition may be formulated as any pharmaceutically useful form, e.g., as an aerosol, a cream, a gel, a gel cap, a pill, a microparticle, a nanoparticle, an injection or infusion solution, a capsule, a tablet, a syrup, a transdermal patch, a subcutaneous patch, a dry powder, an inhalation formulation, in a medical device, suppository, buccal, or sublingual formulation, parenteral formulation, or an ophthalmic solution or suspension.
  • Some dosage forms, such as tablets and capsules are subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.
  • compositions, and methods of manufacturing such compositions, suitable for administration as contemplated herein are known in the art.
  • known techniques include, for example, U.S. Pat. Nos. 4,983,593, 5,013,557, 5,456,923, 5,576,025, 5,723,269, 5,858,411, 6,254,889, 6,303,148, 6,395,302, 6,497,903, 7,060,296, 7,078,057, 7,404,828, 8,202,912, 8,257,741, 8,263,128, 8,337,899, 8,431,159, 9,028,870, 9,060,938, 9,211,261, 9,265,731, 9,358,478, and 9,387,252, incorporated by reference herein.
  • compositions contemplated here can optionally include a carrier.
  • Carriers must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated.
  • the carrier can be inert or it can possess pharmaceutical benefits of its own.
  • the amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.
  • Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, fillers, flavorants, glidents, lubricants, pH modifiers, preservatives, stabilizers, surfactants, solubilizers, tableting agents, and wetting agents.
  • Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others.
  • Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin; talc, and vegetable oils.
  • examples of other matrix materials, fillers, or diluents include lactose, mannitol, xylitol, microcrystalline cellulose, calcium diphosphate, and starch.
  • surface active agents include sodium lauryl sulfate and polysorbate 80.
  • Examples of drug complexing agents or solubilizers include the polyethylene glycols, caffeine, xanthene, gentisic acid and cylodextrins.
  • Examples of disintegrants include sodium starch gycolate, sodium alginate, carboxymethyl cellulose sodium, methyl cellulose, colloidal silicon dioxide, and croscarmellose sodium.
  • Examples of binders include methyl cellulose, microcrystalline cellulose, starch, and gums such as guar gum, and tragacanth.
  • Examples of lubricants include magnesium stearate and calcium stearate.
  • pH modifiers include acids such as citric acid, acetic acid, ascorbic acid, lactic acid, aspartic acid, succinic acid, phosphoric acid, and the like; bases such as sodium acetate, potassium acetate, calcium oxide, magnesium oxide, trisodium phosphate, sodium hydroxide, calcium hydroxide, aluminum hydroxide, and the like, and buffers generally comprising mixtures of acids and the salts of said acids.
  • bases such as sodium acetate, potassium acetate, calcium oxide, magnesium oxide, trisodium phosphate, sodium hydroxide, calcium hydroxide, aluminum hydroxide, and the like, and buffers generally comprising mixtures of acids and the salts of said acids.
  • buffers generally comprising mixtures of acids and the salts of said acids.
  • optionalal other active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the compound of the present invention.
  • the pharmaceutical composition for administration further includes a compound or salt of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII and optionally comprises one or more of a phosphoglyceride; phosphatidylcholine; dipalmitoyl phosphatidylcholine (DPPC); dioleylphosphatidyl ethanolamine (DOPE); dioleyloxypropyltriethylammonium (DOTMA); dioleoylphosphatidylcholine; cholesterol; cholesterol ester; diacylglycerol; diacylglycerolsuccinate; diphosphatidyl glycerol (DP
  • the pharmaceutical preparation may include polymers for controlled delivery of the described compounds, including, but not limited to pluronic polymers, polyesters (e.g., polylactic acid, poly(lactic-co-glycolic acid), polycaprolactone, polyvalerolactone, poly(1,3-dioxan-2one)); polyanhydrides (e.g., poly(sebacic anhydride)); polyethers (e.g., polyethylene glycol); polyurethanes; polymethacrylates; polyacrylates; and polycyanoacrylates.
  • polymers may be modified with polyethylene glycol (PEG), with a carbohydrate, and/or with acyclic polyacetals derived from polysaccharides. See, e.g., Papisov, 2001, ACS Symposium Series, 786:301, incorporated by reference herein.
  • the compounds of the present invention can be formulated as particles.
  • the particles are or include microparticles.
  • the particles are or include nanoparticles.
  • common techniques for preparing particles include, but are not limited to, solvent evaporation, solvent removal, spray drying, phase inversion, coacervation, and low temperature casting. Suitable methods of particle formulation are briefly described below.
  • Pharmaceutically acceptable excipients including pH modifying agents, disintegrants, preservatives, and antioxidants, can optionally be incorporated into the particles during particle formation.
  • the particles are derived through a solvent evaporation method.
  • a compound described herein or polymer matrix and one or more compounds described herein
  • a volatile organic solvent such as methylene chloride.
  • the organic solution containing a compound described herein is then suspended in an aqueous solution that contains a surface active agent such as poly(vinyl alcohol).
  • the resulting emulsion is stirred until most of the organic solvent evaporated, leaving solid nanoparticles or microparticles.
  • the resulting nanoparticles or microparticles are washed with water and dried overnight in a lyophilizer. Nanoparticles with different sizes and morphologies can be obtained by this method.
  • compositions which contain labile polymers may degrade during the fabrication process due to the presence of water.
  • labile polymers such as certain polyanhydrides
  • methods which are performed in completely or substantially anhydrous organic solvents can be used to make the particles.
  • Solvent removal can also be used to prepare particles from a compound that is hydrolytically unstable.
  • the compound or polymer matrix and one or more compounds
  • a volatile organic solvent such as methylene chloride.
  • This mixture is then suspended by stirring in an organic oil (such as silicon oil) to form an emulsion.
  • Solid particles form from the emulsion, which can subsequently be isolated from the supernatant.
  • the external morphology of spheres produced with this technique is highly dependent on the identity of the drug.
  • an active compound as described herein is administered to a patient in need thereof as particles formed by solvent removal.
  • the present invention provides particles formed by solvent removal comprising a compound of the present invention and one or more pharmaceutically acceptable excipients as defined herein.
  • the particles formed by solvent removal comprise a compound of the present invention and an additional therapeutic agent.
  • the particles formed by solvent removal comprise a compound of the present invention, an additional therapeutic agent, and one or more pharmaceutically acceptable excipients.
  • any of the described particles formed by solvent removal can be formulated into a tablet and then coated to form a coated tablet.
  • the particles formed by solvent removal are formulated into a tablet but the tablet is uncoated.
  • the particles are derived by spray drying.
  • a compound or polymer matrix and one or more compounds
  • an organic solvent such as methylene chloride.
  • the solution is pumped through a micronizing nozzle driven by a flow of compressed gas, and the resulting aerosol is suspended in a heated cyclone of air, allowing the solvent to evaporate from the micro droplets, forming particles.
  • Microparticles and nanoparticles can be obtained using this method.
  • an active compound as described herein is administered to a patient in need thereof as a spray dried dispersion (SDD).
  • the present invention provides a spray dried dispersion (SDD) comprising a compound of the present invention and one or more pharmaceutically acceptable excipients as defined herein.
  • the SDD comprises a compound of the present invention and an additional therapeutic agent.
  • the SDD comprises a compound of the present invention, an additional therapeutic agent, and one or more pharmaceutically acceptable excipients.
  • any of the described spray dried dispersions can be coated to form a coated tablet.
  • the spray dried dispersion is formulated into a tablet but is uncoated.
  • Particles can be formed from the active compound as described herein using a phase inversion method.
  • the compound or polymer matrix and one or more active compounds
  • the solution is poured into a strong non-solvent for the compound to spontaneously produce, under favorable conditions, microparticles or nanoparticles.
  • the method can be used to produce nanoparticles in a wide range of sizes, including, for example, from nanoparticles to microparticles, typically possessing a narrow particle size distribution.
  • an active compound as described herein is administered to a patient in need thereof as particles formed by phase inversion.
  • the present invention provides particles formed by phase inversion comprising a compound of the present invention and one or more pharmaceutically acceptable excipients as defined herein.
  • the particles formed by phase inversion comprise a compound of the present invention and an additional therapeutic agent.
  • the particles formed by phase inversion comprise a compound of the present invention, an additional therapeutic agent, and one or more pharmaceutically acceptable excipients.
  • any of the described particles formed by phase inversion can be formulated into a tablet and then coated to form a coated tablet.
  • the particles formed by phase inversion are formulated into a tablet but the tablet is uncoated.
  • Coacervation involves the separation of a compound (or polymer matrix and one or more compounds) solution into two immiscible liquid phases.
  • One phase is a dense coacervate phase, which contains a high concentration of the compound, while the second phase contains a low concentration of the compound.
  • the compound forms nanoscale or microscale droplets, which harden into particles.
  • Coacervation may be induced by a temperature change, addition of a non-solvent or addition of a micro-salt (simple coacervation), or by the addition of another polymer thereby forming an interpolymer complex (complex coacervation).
  • an active compound as described herein is administered to a patient in need thereof as particles formed by coacervation.
  • the present invention provides particles formed by coacervation comprising a compound of the present invention and one or more pharmaceutically acceptable excipients as defined herein.
  • the particles formed by coacervation comprise a compound of the present invention and an additional therapeutic agent.
  • the particles formed by coacervation comprise a compound of the present invention, an additional therapeutic agent, and one or more pharmaceutically acceptable excipients.
  • any of the described particles formed by coacervation can be formulated into a tablet and then coated to form a coated tablet.
  • the particles formed by coacervation are formulated into a tablet but the tablet is uncoated.
  • a compound of the present invention is administered to a patient in need thereof as particles formed by low temperature casting.
  • the present invention provides particles formed by low temperature casting comprising a compound of the present invention and one or more pharmaceutically acceptable excipients as defined herein.
  • the particles formed by low temperature casting comprise a compound of the present invention and an additional therapeutic agent.
  • the particles formed by low temperature casting comprise a compound of the present invention, an additional therapeutic agent, and one or more pharmaceutically acceptable excipients.
  • any of the described particles formed by low temperature casting can be formulated into a tablet and then coated to form a coated tablet.
  • the particles formed by low temperature casting are formulated into a tablet but the tablet is uncoated.
  • an effective amount of an active compound as described herein is incorporated into a nanoparticle, e.g. for convenience of delivery and/or extended release delivery.
  • a nanoparticle e.g. for convenience of delivery and/or extended release delivery.
  • the use of materials in nanoscale provides one the ability to modify fundamental physical properties such as solubility, diffusivity, blood circulation half-life, drug release characteristics, and/or immunogenicity.
  • a number of nanoparticle-based therapeutic and diagnostic agents have been developed for the treatment of cancer, diabetes, pain, asthma, allergy, and infections. These nanoscale agents may provide more effective and/or more convenient routes of administration, lower therapeutic toxicity, extend the product life cycle, and ultimately reduce health-care costs. As therapeutic delivery systems, nanoparticles can allow targeted delivery and controlled release.
  • nanoparticle-based compound delivery can be used to release compounds at a sustained rate and thus lower the frequency of administration, deliver drugs in a targeted manner to minimize systemic side effects, or deliver two or more drugs simultaneously for combination therapy to generate a synergistic effect and suppress drug resistance.
  • a number of nanotechnology-based therapeutic products have been approved for clinical use. Among these products, liposomal drugs and polymer-based conjugates account for a large proportion of the products. See, Zhang, L., et al., Nanoparticles in Medicine: Therapeutic Applications and Developments, Clin. Pharm. and Ther., 83(5):761-769, 2008.
  • polyesters examples include poly(L-lactide-co-L-lysine) (Barrera et al., 1993, J. Am. Chem. Soc., 115:11010), poly(serine ester) (Zhou et al., 1990, Macromolecules, 23:3399), poly(4-hydroxy-L-proline ester) (Putnam et al., 1999, Macromolecules, 32:3658; and Lim et al., 1999, J. Am. Chem. Soc., 121:5633), and poly(4-hydroxy-L-proline ester) (Putnam et al., 1999, Macromolecules, 32:3658; and Lim et al., 1999, J.
  • the polymeric particle is between about 0.1 nm to about 10000 nm, between about 1 nm to about 1000 nm, between about 10 nm and 1000 nm, between about 1 and 100 nm, between about 1 and 10 nm, between about 1 and 50 nm, between about 100 nm and 800 nm, between about 400 nm and 600 nm, or about 500 nm.
  • the microparticles are no more than about 0.1 nm, 0.5 nm, 1.0 nm, 5.0 nm, 10 nm, 25 nm, 50 nm, 75 nm, 100 nm, 150 nm, 200 nm, 250 nm, 300 nm, 400 nm, 450 nm, 500 nm, 550 nm, 600 nm, 650 nm, 700 nm, 750 nm, 800 nm, 850 nm, 900 nm, 950 nm, 1000 nm, 1250 nm, 1500 nm, 1750 nm, or 2000 nm.
  • a compound described herein may be covalently coupled to a polymer used in the nanoparticle, for example a polystyrene particle, PLGA particle, PLA particle, or other nanoparticle.
  • compositions can be formulated for oral administration. These compositions can contain any amount of active compound that achieves the desired result, for example between 0.1 and 99 weight % (wt. %) of the compound and usually at least about 5 wt. % of the compound. Some embodiments contain at least about 10%, 15%, 20%, 25 wt. % to about 50 wt. % or from about 5 wt. % to about 75 wt. % of the compound.
  • compositions suitable for rectal administration are typically presented as unit dose suppositories. These may be prepared by admixing the active compound with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • conventional solid carriers for example, cocoa butter
  • compositions suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil.
  • Carriers which may be used include petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof.
  • compositions suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • Pharmaceutical compositions suitable for transdermal administration may also be delivered by iontophoresis (see, for example, Pharmaceutical Research 3 (6):318 (1986)) and typically take the form of an optionally buffered aqueous solution of the active compound.
  • microneedle patches or devices are provided for delivery of drugs across or into biological tissue, particularly the skin. The microneedle patches or devices permit drug delivery at clinically relevant rates across or into skin or other tissue barriers, with minimal or no damage, pain, or irritation to the tissue.
  • compositions suitable for administration to the lungs can be delivered by a wide range of passive breath driven and active power driven single/-multiple dose dry powder inhalers (DPI).
  • DPI dry powder inhalers
  • the devices most commonly used for respiratory delivery include nebulizers, metered-dose inhalers, and dry powder inhalers.
  • nebulizers include jet nebulizers, ultrasonic nebulizers, and vibrating mesh nebulizers.
  • Selection of a suitable lung delivery device depends on parameters, such as nature of the drug and its formulation, the site of action, and pathophysiology of the lung.
  • inhalation drug delivery devices and methods include, for example, U.S. Pat. No. 7,383,837 titled “Inhalation device” (SmithKline Beecham Corporation); WO/2006/033584 titled “Powder inhaler” (Glaxo SmithKline Pharmaceuticals SA); WO/2005/044186 titled “Inhalable pharmaceutical formulations employing desiccating agents and methods of administering the same” (Glaxo Group Ltd and SmithKline Beecham Corporation); U.S. Pat. No. 9,095,670 titled “Inhalation device and method of dispensing medicament”, U.S. Pat. No.
  • WO/2010/009087 titled “Iontophoretic Delivery of a Controlled-Release Formulation in the Eye”, (Liquidia Technologies, Inc. and Eyegate Pharmaceuticals, Inc.) and WO/2009/132206 titled “Compositions and Methods for Intracellular Delivery and Release of Cargo”, WO/2007/133808 titled “Nano-particles for cosmetic applications”, WO/2007/056561 titled “Medical device, materials, and methods”, WO/2010/065748 titled “Method for producing patterned materials”, WO/2007/081876 titled “Nanostructured surfaces for biomedical/biomaterial applications and processes thereof” (Liquidia Technologies, Inc.).
  • Additional non-limiting examples of methods and devices for drug delivery to the eye include, for example, WO2011/106702 and U.S. Pat. No. 8,889,193 titled “Sustained delivery of therapeutic agents to an eye compartment”, WO2013/138343 and U.S. Pat. No. 8,962,577 titled “Controlled release formulations for the delivery of HIF-1 inhibitors”, WO/2013/138346 and US2013/0272994 titled “Non-Linear Multiblock Copolymer-Drug Conjugates for the Delivery of Active Agents”, WO2005/072710 and U.S. Pat. No.
  • drug delivery devices and methods include, for example, US20090203709 titled “Pharmaceutical Dosage Form For Oral Administration Of Tyrosine Kinase Inhibitor” (Abbott Laboratories); US20050009910 titled “Delivery of an active drug to the posterior part of the eye via subconjunctival or periocular delivery of a prodrug”, US 20130071349 titled “Biodegradable polymers for lowering intraocular pressure”, U.S. Pat. No. 8,481,069 titled “Tyrosine kinase microspheres”, U.S. Pat. No.
  • an effective amount of an active compound or its salt or composition as described herein is used to treat a medical disorder which is an inflammatory or immune condition, a disorder mediated by the complement cascade (including a dysfunctional cascade) including a complement-mediated disease or disorder including a complement factor D-related disorder or alternative complement pathway-related disorder, a disorder or abnormality of a cell that adversely affects the ability of the cell to engage in or respond to normal complement activity, or an undesired complement-mediated response to a medical treatment, such as surgery or other medical procedure or a pharmaceutical or biopharmaceutical drug administration, a blood transfusion, or other allogenic tissue or fluid administration.
  • a medical disorder which is an inflammatory or immune condition
  • a disorder mediated by the complement cascade including a dysfunctional cascade
  • a complement-mediated disease or disorder including a complement factor D-related disorder or alternative complement pathway-related disorder a disorder or abnormality of a cell that adversely affects the ability of the cell to engage in or respond to normal complement activity, or an undesired
  • a complement-mediated disease or disorder is a disease or disorder in which the amount or activity of complement is such as to cause disease or disorder in an individual.
  • the complement-mediated disease or disorder is selected from the group consisting of autoimmune disease, cancer, hematological disease, infectious disease, inflammatory disease, ischemia-reperfusion injury, neurodegenerative disease, neurodegenerative disorder, ocular disease, renal disease, transplant rejection, vascular disease, and vasculitis disease.
  • the complement-mediated disease or disorder is an autoimmune disease.
  • the complement-mediated disease or disorder is cancer.
  • the complement-mediated disease or disorder is an infectious disease.
  • the complement-mediated disease or disorder is an inflammatory disease.
  • the complement-mediated disease or disorder is a hematological disease. In some embodiments, the complement-mediated disease or disorder is an ischemia-reperfusion injury. In some embodiments, the complement-mediated disease or disorder is ocular disease. In some embodiments, the complement-mediated disease or disorder is a renal disease. In some embodiments, the complement-mediated disease or disorder is transplant rejection. In some embodiments, the complement-mediated disease or disorder is antibody-mediated transplant rejection. In some embodiments, the complement-mediated disease or disorder is a vascular disease. In some embodiments, the complement-mediated disease or disorder is a vasculitis disorder. In some embodiments, the complement-mediated disease or disorder is a neurodegenerative disease or disorder. In some embodiments, the complement-mediated disease is a neurodegenerative disease. In some embodiments, the complement-mediated disorder is a neurodegenerative disorder. In some embodiments, the complement-mediated disorder is a neurodegenerative disorder. In some embodiments, the complement-mediated disease or disorder is a tauopathy.
  • a method for the treatment of C3 glomerulonephritis includes the administration of an effective amount of a compound to a host of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII, or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition.
  • a method for the treatment of paroxysmal nocturnal hemoglobinuria includes the administration of an effective amount of a compound to a host of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII, or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition.
  • PNH paroxysmal nocturnal hemoglobinuria
  • a method for the treatment of wet or dry age-related macular degeneration (AMD) in a host includes the administration of an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII, or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition.
  • ALD age-related macular degeneration
  • a method for the treatment of rheumatoid arthritis in a host includes the administration of an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII, or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition.
  • a method for the treatment of multiple sclerosis in a host includes the administration of an effective amount of a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, or Formula XXIII, or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition.
  • the active compound or its pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition, as disclosed herein is also useful for administration in combination (in the same or a different dosage form) or alternation with a second pharmaceutical agent for use in ameliorating or reducing a side effect of the second pharmaceutical agent.
  • the active compound may be used in combination with an adoptive cell transfer therapy to reduce an inflammatory response associated with such therapy, for example, a cytokine mediated response such as cytokine response syndrome.
  • the adoptive cell transfer therapy is a chimeric antigen receptor T-Cell (CAR T) or a dendritic cell used to treat a hematologic or solid tumor, for example, a B-cell related hematologic cancer.
  • the hematologic or solid tumor is acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), non-Hodgkin's lymphoma, chronic lymphocytic leukemia (CLL), pancreatic cancer, glioblastoma, or a cancer that expresses CD19.
  • the associated inflammatory response is a cytokine mediated response.
  • Another embodiment includes the administration of an effective amount of an active compound or a pharmaceutically acceptable salt, prodrug, isotopic analog, N-oxide, or isolated isomer thereof, optionally in a pharmaceutically acceptable composition to a host to treat an ocular, pulmonary, gastrointestinal, or other disorder that can benefit from topical or local delivery.
  • any of the compounds described herein can be administered to the eye in any desired form of administration, including via intravitreal, intrastromal, intracameral, sub-tenon, sub-retinal, retro-bulbar, peribulbar, suprachorodial, choroidal, subchoroidal, conjunctival, subconjunctival, episcleral, posterior juxtascleral, scleral, circumcorneal, and tear duct injections, or through a mucus, mucin, or a mucosal barrier, in an immediate or controlled release fashion.
  • intravitreal intrastromal, intracameral, sub-tenon, sub-retinal, retro-bulbar, peribulbar, suprachorodial, choroidal, subchoroidal, conjunctival, subconjunctival, episcleral, posterior juxtascleral, scleral, circumcorneal, and tear duct injections, or through a mucus, mucin, or a mucos
  • the active compound includes a lipophilic group, such as a lipophilic acyl group, which is delivered to the eye in a polymeric drug delivery system such as polylactic acid, polylactide-co-glycolide, polyglycolide or other erodible polymer, or a combination thereof, or in another type of lipophilic material for ocular delivery.
  • a lipophilic active molecule is more soluble in the polymeric or other form of delivery system than in ocular fluid.
  • an active compound provided herein can be used to treat or prevent a disorder in a host mediated by complement factor D, or by an excessive or detrimental amount of the complement-C3 amplification loop of the complement pathway.
  • the invention includes methods to treat or prevent complement associated disorders that are induced by antibody-antigen interactions, a component of an immune or autoimmune disorder or by ischemic injury.
  • the invention also provides methods to decrease inflammation or an immune response, including an autoimmune response, where mediated or affected by factor D.
  • the disorder is selected from fatty liver and conditions stemming from fatty liver, such as nonalcoholic steatohepatitis (NASH), liver inflammation, cirrhosis and liver failure.
  • NASH nonalcoholic steatohepatitis
  • a method is provided for treating fatty liver disease in a host by administering an effective amount of an active compound or its salt or composition as described herein.
  • an active compound or its salt or composition as described herein is used to modulate an immune response prior to or during surgery or other medical procedure.
  • One non-limiting example is use in connection with acute or chronic graft versus host disease, which is a common complication as a result of allogeneic tissue transplant, and can also occur as a result of a blood transfusion.
  • the present invention provides a method of treating or preventing dermatomyositis by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • the present invention provides a method of treating or preventing amyotrophic lateral sclerosis by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • the present invention provides a method of treating or preventing abdominal aortic aneurysm, hemodialysis complications, hemolytic anemia, or hemodialysis by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • a method for the treatment or prevention of cytokine or inflammatory reactions in response to the administration of pharmaceutical or biotherapeutic (e.g. CAR T-cell therapy or monoclonal antibody therapy) in a host by administering an effective amount of an active compound or its salt or composition as described herein.
  • cytokine or inflammatory reactions may occur in response to a number of factors, such as the administrations of biotherapeutics.
  • the cytokine or inflammatory reaction is cytokine release syndrome.
  • the cytokine or inflammatory reaction is tumor lysis syndrome (which also leads to cytokine release). Symptoms of cytokine release syndrome range from fever, headache, and skin rashes to bronchospasm, hypotension and even cardiac arrest. Severe cytokine release syndrome is described as cytokine storm, and can be fatal.
  • Fatal cytokine storms have been observed in response to infusion with several monoclonal antibody therapeutics. See, Abramowicz D, et al. “Release of tumor necrosis factor, interleukin-2, and gamma-interferon in serum after injection of OKT3 monoclonal antibody in kidney transplant recipients” Transplantation (1989) 47(4):606-8; Chatenoud L, et al. “In vivo cell activation following OKT3 administration. Systemic cytokine release and modulation by corticosteroids” Transplantation (1990) 49(4):697-702; and Lim L C, Koh L P, and Tan P.
  • bi-specific T-cell engagers directs T-cells to target and bind with a specific antigen on the surface of a cancer cell.
  • Blinatumomab (Amgen)
  • Amgen a BiTE has recently been approved as a second line therapy in Philadelphia chromosome-negative relapsed or refractory acute lymphoblastic leukemia.
  • Blinatumomab is given by continuous intravenous infusion in 4-week cycles.
  • the use of BiTE agents has been associated with adverse immune responses, including cytokine release syndrome.
  • cytokines in the CRS associated with ACT include IL-10, IL-6, and IFN- ⁇ (Klinger et al., Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific BiTE antibody blinatumomab. Blood (2012) 119:6226-6233).
  • the disorder is episcleritis, idiopathic episcleritis, anterior episcleritis, or posterior episcleritis.
  • the disorder is idiopathic anterior uveitis, HLA-B27 related uveitis, herpetic keratouveitis, Posner Schlossman syndrome, Fuch's heterochromic iridocyclitis, or cytomegalovirus anterior uveitis.
  • the present invention provides a method of treating or preventing a C3 glomurenopathy by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • the disorder is selected from dense deposit disease (DDD) and C3 glomerulonephritis (C3GN).
  • the present invention provides a method of treating or preventing a IC-MPGN by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • the present invention provides a method of treating or preventing a paroxysmal nocturnal hemoglobinuria (PNH) by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • PNH paroxysmal nocturnal hemoglobinuria
  • the present invention provides a method of treating or preventing age-related macular degeneration (AMD) by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • AMD age-related macular degeneration
  • the present invention provides a method of treating or preventing rheumatoid arthritis by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • the present invention provides a method of treating or preventing multiple sclerosis by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • the present invention provides a method of treating or preventing myasthenia gravis by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • the present invention provides a method of treating or preventing atypical hemolytic uremic syndrome (aHUS) by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • aHUS atypical hemolytic uremic syndrome
  • the present invention provides a method of treating or preventing neuromyelitis optica (NMO) by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein.
  • NMO neuromyelitis optica
  • the present invention provides a method of treating or preventing a disorder as described below by administering to a subject in need thereof an effective amount of an active compound or its salt or composition as described herein, including: vitritis, sarcoidosis, syphilis, tuberculosis, or Lyme disease; retinal vasculitis, Eales disease, tuberculosis, syphilis, or toxoplasmosis; neuroretinitis, viral retinitis, or acute retinal necrosis; varicella zoster virus, herpes simplex virus, cytomegalovirus, Epstein-Barr virus, lichen planus, or Dengue-associated disease (e.g., hemorraghic Dengue Fever); Masquerade syndrome, contact dermatitis, trauma induced inflammation, UVB induced inflammation, eczema, granuloma annulare, or acne.
  • an active compound or its salt or composition as described herein, including: vitritis, sarcoidos
  • the disorder is selected from: acute myocardial infarction, aneurysm, cardiopulmonary bypass, dilated cardiomyopathy, complement activation during cardiopulmonary bypass operations, coronary artery disease, restenosis following stent placement, or percutaneous transluminal coronary angioplasty (PTCA); antibody-mediated transplant rejection, anaphylactic shock, anaphylaxis, allogenic transplant, humoral and vascular transplant rejection, graft dysfunction, graft-versus-host disease, Graves' disease, adverse drug reactions, or chronic graft vasculopathy; allergic bronchopulmonary aspergillosis, allergic neuritis, drug allergy, radiation-induced lung injury, eosinophilic pneumonia, radiographic contrast media allergy, bronchiolitis obliterans, or interstitial pneumonia; parkinsonism-dementia complex, sporadic frontotemporal dementia, frontotemporal dementia with Parkinsonism linked to chromosome 17, frontotemporal lobar degeneration, tangle only dementia, cerebral am
  • the disorder is selected from: atopic dermatitis, dermatitis, dermatomyositis bullous pemphigoid, scleroderma, sclerodermatomyositis, psoriatic arthritis, pemphigus vulgaris, Discoid lupus erythematosus, cutaneous lupus, chilblain lupus erythematosus, or lupus erythematosus-lichen planus overlap syndrome; cryoglobulinemic vasculitis, mesenteric/enteric vascular disorder, peripheral vascular disorder, antineutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV), IL-2 induced vascular leakage syndrome, or immune complex vasculitis; angioedema, low platelets (HELLP) syndrome, sickle cell disease, platelet refractoriness, red cell casts, or typical or infectious hemolytic uremic syndrome (tHUS); hematuri
  • the disorder is selected from: wet (exudative) AMD, dry (non-exudative) AMD, chorioretinal degeneration, choroidal neovascularization (CNV), choroiditis, loss of RPE function, loss of vision (including loss of visual acuity or visual field), loss of vision from AMD, retinal damage in response to light exposure, retinal degeneration, retinal detachment, retinal dysfunction, retinal neovascularization (RNV), retinopathy of prematurity, pathological myopia, or RPE degeneration; pseudophakic bullous keratopathy, symptomatic macular degeneration related disorder, optic nerve degeneration, photoreceptor degeneration, cone degeneration, loss of photoreceptor cells, pars planitis, scleritis, proliferative vitreoretinopathy, or formation of ocular drusen; chronic urticaria, Churg-Strauss syndrome, cold agglutinin disease (CAD), cortico
  • the disorder is selected from: hyperlipidemia, hypertension, hypoalbuminemia, hypobolemic shock, hypocomplementemic urticarial vasculitis syndrome, hypophosphastasis, hypovolemic shock, idiopathic pneumonia syndrome, or idiopathic pulmonary fibrosis; inclusion body myositis, intestinal ischemia, iridocyclitis, ulceris, juvenile chronic arthritis, Kawasaki's disease (arteritis), or lipiduria; membranoproliferative glomerulonephritis (MPGN) I, microscopic polyangiitis, mixed cryoglobulinemia, molybdenum cofactor deficiency (MoCD) type A, pancreatitis, panniculitis, Pick's disease, polyarteritis nodosa (PAN), progressive subcortical gliosis, proteinuria, reduced glomerular filtration rate (GFR), or renovascular disorder; multiple organ failure, multiple system atrophy
  • an active compound or its salt or composition as described herein is useful for treating or preventing a disorder selected from autoimmune oophoritis, endometriosis, autoimmune orchitis, Ord's thyroiditis, autoimmune enteropathy, coeliac disease, Hashimoto's encephalopathy, antiphospholipid syndrome (APLS) (Hughes syndrome), aplastic anemia, autoimmune lymphoproliferative syndrome (Canale-Smith syndrome), autoimmune neutropenia, Evans syndrome, pernicious anemia, pure red cell aplasia, thrombocytopenia, adipose dolorosa (Dercum's disease), adult onset Still's disease, ankylosing spondylitis, CREST syndrome, drug-induced lupus, eosinophilic fasciitis (Shulman's syndrome), Felty syndrome, IgG4-related disease, mixed connective tissue disease (MCTD), palindromic rheumatism (Hench-R
  • eye disorders that may be treated according to the compositions and methods disclosed herein include amoebic keratitis, fungal keratitis, bacterial keratitis, viral keratitis, onchorcercal keratitis, bacterial keratoconjunctivitis, viral keratoconjunctivitis, corneal dystrophic diseases, Fuchs' endothelial dystrophy, Sjogren's syndrome, Stevens-Johnson syndrome, autoimmune dry eye diseases, environmental dry eye diseases, corneal neovascularization diseases, post-corneal transplant rejection prophylaxis and treatment, autoimmune uveitis, infectious uveitis, posterior uveitis (including toxoplasmosis), pan-uveitis, an inflammatory disease of the vitreous or retina, endophthalmitis prophylaxis and treatment, macular edema, macular degeneration, age related macular degeneration, proliferative and non-proliferative diabetic retin
  • the disorder is selected from glaucoma, diabetic retinopathy, blistering cutaneous diseases (including bullous pemphigoid, pemphigus, and epidermolysis bullosa), ocular cicatrical pemphigoid, uveitis, adult macular degeneration, diabetic retinopa retinitis pigmentosa, macular edema, diabetic macular edema, Behcet's uveitis, multifocal choroiditis, Vogt-Koyangi-Harada syndrome, imtermediate uveitis, birdshot retino-chorioditis, sympathetic ophthalmia, ocular dicatricial pemphigoid, ocular pemphigus, nonartertic ischemic optic neuropathy, postoperative inflammation, and retinal vein occlusion, or central retinal vein occulusion (CVRO).
  • glaucoma including bullous pemphigo
  • complement mediated diseases include ophthalmic diseases (including early or neovascular age-related macular degeneration and geographic atrophy), autoimmune diseases (including arthritis, rheumatoid arthritis), respiratory diseases, cardiovascular diseases.
  • ophthalmic diseases including early or neovascular age-related macular degeneration and geographic atrophy
  • autoimmune diseases including arthritis, rheumatoid arthritis
  • respiratory diseases including cardiovascular diseases.
  • the compounds of the invention are suitable for use in the treatment of diseases and disorders associated with fatty acid metabolism, including obesity and other metabolic disorders.
  • disorders that may be treated or prevented by an active compound or its salt or composition as described herein also include, but are not limited to: hereditary angioedema, capillary leak syndrome, hemolytic uremic syndrome (HUS), neurological disorders, Guillain Barre Syndrome, diseases of the central nervous system and other neurodegenerative conditions, glomerulonephritis (including membrane proliferative glomerulonephritis), SLE nephritis, proliferative nephritis, liver fibrosis, tissue regeneration and neural regeneration, or Barraquer-Simons Syndrome; inflammatory effects of sepsis, systemic inflammatory response syndrome (SIRS), disorders of inappropriate or undesirable complement activation, interleukin-2 induced toxicity during IL-2 therapy, inflammatory disorders, inflammation of autoimmune diseases, system lupus erythematosus (SLE), lupus nephritides, arthritis, immune complex disorders and autoimmune diseases, systemic lupus, or lupus ery
  • a method for the treatment of sickle cell in a host includes the administration of an effective amount of an active compound or its salt or composition as described herein.
  • a method for the treatment of immunothrombocytopenic purpura (ITP), thrombotic thrombocytopenic purpura (TTP), or idiopathic thrombocytopenic purpura (ITP) in a host includes the administration of an effective amount of an active compound or its salt or composition as described herein.
  • a method for the treatment of ANCA-vasculitis in a host includes the administration of an effective amount of an active compound or its salt or composition as described herein.
  • a method for the treatment of IgA nephropathy in a host includes the administration of an effective amount of an active compound or its salt or composition as described herein.
  • a method for the treatment of rapidly progressing glomerulonephritis (RPGN) in a host is provided that includes the administration of an effective amount of an active compound or its salt or composition as described herein.
  • RPGN rapidly progressing glomerulonephritis
  • a method for the treatment of lupus nephritis in a host is provided that includes the administration of an effective amount of an active compound or its salt or composition as described herein.
  • a method for the treatment of hemorraghic dengue fever, in a host is provided that includes the administration of an effective amount of an active compound or its salt or composition as described herein.
  • an active compound or its salt or composition as described herein is used in the treatment of an autoimmune disorder.
  • the complement pathway enhances the ability of antibodies and phagocytic cells to clear microbes and damaged cells from the body. It is part of the innate immune system and in healthy individuals is an essential process. Inhibiting the complement pathway will decrease the body's immune system response. Therefore, it is an object of the present invention to treat autoimmune disorders by administering an effective does of an active compound or its salt or composition as described herein to a subject in need thereof.
  • the autoimmune disorder is caused by activity of the complement system. In some embodiments the autoimmune disorder is caused by activity of the alternative complement pathway. In some embodiments the autoimmune disorder is caused by activity of the classical complement pathway. In another embodiment the autoimmune disorder is caused by a mechanism of action that is not directly related to the complement system, such as the over-proliferation of T-lymphocytes or the over-production of cytokines.
  • Non-limiting examples of autoimmune disorders include: lupus, allograft rejection, autoimmune thyroid diseases (such as Graves' disease and Hashimoto's thyroiditis), autoimmune uveoretinitis, giant cell arteritis, inflammatory bowel diseases (including Crohn's disease, ulcerative colitis, regional enteritis, granulomatous enteritis, distal ileitis, regional ileitis, and terminal ileitis), diabetes, multiple sclerosis, pernicious anemia, psoriasis, rheumatoid arthritis, sarcoidosis, and scleroderma.
  • autoimmune thyroid diseases such as Graves' disease and Hashimoto's thyroiditis
  • autoimmune uveoretinitis giant cell arteritis
  • inflammatory bowel diseases including Crohn's disease, ulcerative colitis, regional enteritis, granulomatous enteritis, distal ileitis, regional ileitis, and terminal ileitis
  • diabetes
  • an active compound or its salt or composition as described herein is used in the treatment of lupus.
  • lupus include lupus erythematosus, cutaneous lupus, discoid lupus erythematosus, chilblain lupus erythematosus, or lupus erythematosus-lichen planus overlap syndrome.
  • Lupus erythematosus is a general category of disease that includes both systemic and cutaneous disorders.
  • the systemic form of the disease can have cutaneous as well as systemic manifestations.
  • SLE is an inflammatory disorder of unknown etiology that occurs predominantly in women, and is characterized by articular symptoms, butterfly erythema, recurrent pleurisy, pericarditis, generalized adenopathy, splenomegaly, as well as CNS involvement and progressive renal failure.
  • the sera of most patients (over 98%) contain antinuclear antibodies, including anti-DNA antibodies. High titers of anti-DNA antibodies are essentially specific for SLE. Conventional treatment for this disease has been the administration of corticosteroids or immunosuppressants.
  • DLE chronic cutaneous lupus
  • subacute cutaneous lupus subacute cutaneous lupus
  • acute cutaneous lupus a disfiguring chronic disorder primarily affecting the skin with sharply circumscribed macules and plaques that display erythema, follicular plugging, scales, telangiectasia and atrophy. The condition is often precipitated by sun exposure, and the early lesions are erythematous, round scaling papules that are 5 to 10 mm in diameter and display follicular plugging.
  • DLE lesions appear most commonly on the cheeks, nose, scalp, and ears, but they may also be generalized over the upper portion of the trunk, extensor surfaces of the extremities, and on the mucous membranes of the mouth. If left untreated, the central lesion atrophies and leaves a scar. Unlike SLE, antibodies against double-stranded DNA (e.g., DNA-binding test) are almost invariably absent in DLE.
  • MS multiple Sclerosis is an autoimmune demyelinating disorder that is believed to be T lymphocyte dependent.
  • MS generally exhibits a relapsing-remitting course or a chronic progressive course.
  • the etiology of MS is unknown, however, viral infections, genetic predisposition, environment, and autoimmunity all appear to contribute to the disorder.
  • Lesions in MS patients contain infiltrates of predominantly T lymphocyte mediated microglial cells and infiltrating macrophages.
  • CD4+ T lymphocytes are the predominant cell type present at these lesions.
  • the hallmark of the MS lesion is plaque, an area of demyelination sharply demarcated from the usual white matter seen in MRI scans. Histological appearance of MS plaques varies with different stages of the disease.
  • Diabetes can refer to either type 1 or type 2 diabetes.
  • an active compound or its salt or composition as described herein is provided at an effective dose to treat a patient with type 1 diabetes.
  • an active compound or its salt or composition as described herein is provided at an effective dose to treat a patient with type 2 diabetes.
  • Type 1 diabetes is an autoimmune disease. An autoimmune disease results when the body's system for fighting infection (the immune system) attacks a part of the body. In the case of diabetes type 1, the pancreas then produces little or no insulin.
  • an effective amount of an active compound described herein, or it pharmaceutically acceptable salt is used to treat a medical disorder of the central nervous system (CNS) or peripheral nervous system disorders involving complement activation.
  • the CNS disorder is an acquired brain or spinal cord injury, including, but not limited to ischaemic-reperfusion injury or stroke, traumatic brain injury (TBI) and spinal cord injury (SCI).
  • TBI traumatic brain injury
  • SCI spinal cord injury
  • the disorder is a neurodegeneration disorder.
  • the disorder is a neuroinflammation disorder.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat Alzheimer's disease (AD).
  • AD is characterized by two hallmark pathologies; amyloid- ⁇ (A3) plaques and neurofibrillary tangles comprising hyperphosphorylated tau.
  • SNPs single nucleotide polymorphisms
  • CLU complement proteins Clusterin
  • CR1 CR1
  • an effective amount of active compound described herein, or it pharmaceutically acceptable salt is used to treat certain forms of frontotemporal dementia including, but not limited to, Pick's disease, sporadic Frontotemporal dementia and Frontotemporal dementia with Parkinsonism linked to chromosome 17, Progressive supranuclear palsy (PSP), Corticobasal degeneration (CBD), and Subacute sclerosing panencephalitis.
  • frontotemporal dementia including, but not limited to, Pick's disease, sporadic Frontotemporal dementia and Frontotemporal dementia with Parkinsonism linked to chromosome 17, Progressive supranuclear palsy (PSP), Corticobasal degeneration (CBD), and Subacute sclerosing panencephalitis.
  • an effective amount of active compound described herein, or it pharmaceutically acceptable salt is used to treat multiple sclerosis (MS).
  • MS multiple sclerosis
  • C3 has been shown to be deposited in the brains of MS patients.
  • TCC has been shown to be in association with capillary endothelial cells, predominantly within plaques and adjacent white matter. Localization of C activation to areas of active myelin destruction has also been shown, with TCC deposited exclusively in such areas.
  • C3d has been shown to be deposited in association with short segments of disrupted myelin in plaques with low-grade active demyelination and provides evidence for a C contribution to disease progression as well as acute inflammation. See Ingram et al., Complement in multiple sclerosis: its role in disease and potential as a biomarker. Clin Exp Immunol. 2009 February; 155(2):128-39.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat neuromyelitis optica (NMO).
  • NMO Neuromyelitis optica
  • MS neuromyelitis optica
  • IgG NMO-immunoglobulin G
  • NMO patients have higher levels of C3a and anti-C1q antibodies than healthy controls.
  • C3a levels correlated with disease activity, neurological disability and aquaporin-4 IgG. Nytrova et al., Complement activation in patients with neuromyelitis optica. J Neuroimmunol. 2014 Sep. 15; 274(1-2):185-91.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat amyotrophic lateral sclerosis (ALS).
  • ALS is caused by progressive loss of upper and lower (a) motor neurons resulting in denervation of neuromuscular junctions in the peripheral nervous system, progressive muscle weakness, atrophy, spasticity, respiratory failure, and ultimately paralysis and death.
  • Recent studies have shown increased C1q protein in motor cortex and spinal cord of ALS post-mortem tissue; C3 activation fragments and TCC in areas of pathology; C4d and TCC staining of degenerating neurons and glia in ALS motor cortex and spinal cord, and C5aR1 upregulation in areas of pathology.
  • C3d and C4d have been found on oligodendroglia and degenerating neurites, surrounded by CR4-positive microglia, in spinal cord and motor cortex, and C1q, C3, and TCC have been shown to be present on motor end-plates in intercostal muscles in ALS donors even early in the disease process. See Carpanini et al., Therapeutic Inhibition of the Complement System in Diseases of the Central Nervous System, Front. Immunol., 4 Mar. 2019.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat Parkinson's disease (PD).
  • PD is characterized by loss of dopaminergic neurons in the substantia nigra and deposits of the protein ⁇ -synuclein that form the pathological hallmarks of the disease, Lewy bodies. Patients present with resting tremor, bradykinesia, and rigidity. Complement activation has been associated with ⁇ -synuclein and Lewy bodies in Parkinson's disease; in vitro studies have demonstrated that the disease-associated splice variant ⁇ -synuclein 112, but not the full-length protein, cause activation of complement.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat Huntington's disease (HD).
  • HD is an autosomal dominant, inherited neurodegenerative disease characterized by progressive motor symptoms, psychiatric disturbances, and dementia. It is caused by expansion of a three-base-pair (CAG) repeat (39-121 repeats vs. normal range 8-39 repeats) in exon 1 of the HTT gene that translates into a polyglutamine tract at the N-terminus of the protein. This results in a polyglutamine length-dependent misfolding and accumulation of huntingtin protein in the striatum and cortex (layers 3, 5, and 6) followed by neuronal loss in these areas which spreads to the hippocampus.
  • CAG three-base-pair
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat argyrophilic grain dementia, British type amyloid angiopathy, cerebral amyloid angiopathy, Creutzfeldt-Jakob disease, dementia pugilistica, diffuse neurofibrillary tangles with calcification, Down's syndrome, frontotemporal lobar degeneration, Gerstmann-Straussler-Scheinker disease, Hallervorden-Spatz disease, inclusion body myositis, multiple system atrophy (MSA), myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian motor neuron disease with neurofibrillary tangles, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, Tangle only dementia, multi-infarct dementia, ischemic stroke, chronic traumatic traumatic a
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat a hereditary motor and sensory neuropathy (HMSN).
  • the hereditary and sensory neuropathy is Charcot-Marie-Tooth (CMT) disease.
  • the HSMN is Charcot-Marie-Tooth disease type 1A or type 1B.
  • the HSMN is Charcot-Marie-Tooth disease type 2.
  • the HSMN is Dejerine-Sottas disease (Charcot-Marie-Tooth type 3).
  • the HSMN is Refsum disease.
  • the HSMN is Charcot-Marie-Tooth with pyramidal features.
  • the HSMN is Charcot-Marie-Tooth type 6.
  • the HSMN is HMSN+retinitis pigmentosa.
  • an active compound as described herein is used to treat Churg-Strauss syndrome.
  • an active compound as described herein is used to treat a peripheral artery disease (PAD).
  • PAD peripheral artery disease
  • an effective amount of active compound described herein, or it pharmaceutically acceptable salt to treat myasthenia gravis with CNS involvement is provided.
  • an effective amount of active compound described herein, or it pharmaceutically acceptable salt is used to treat dementia with Lewy bodies.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat an individual suffering from prion disease.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat Behcet's Disease.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat congenital myasthenia.
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat subacute sclerosing panencephalitis (SSPE).
  • SSPE subacute sclerosing panencephalitis
  • active compound described herein, or it pharmaceutically acceptable salt is used to treat Guillain-Barre syndrome.
  • the CNS disorder to be treated is a demyelinating disease, including, but not limited to demyelinating myelinoclastic diseases and demyelinating leukostrophic disease.
  • the disorder to be treated is a demyelinating myelonoclastic disease including, but not limited to, multiple sclerosis, neuromyelitis optica, neuromyelitis optica spectrum of disorders (NMOSD), idiopathic inflammatory demyelinating diseases (IIDD), anti-NMDA receptor encephalitis, acute disseminated encephalomyelitis, anti-MOG autoimmune encephalomyelitis, chronic relapsing inflammatory optic neuritis (CRION), acute disseminated encephalomyelitis (ADEM), immune-mediated encephalomyelitis, progressive multifocal leukoencephalopathy (PML); McDonalds-positive multiple sclerosis, acute hemorrhagic leukoencephalitis, Rasmussen's Encephalitis, Marburg multiple sclerosis, pseudotumefactive and tumefactive multiple sclerosis, Balo concentric sclerosis, diffuse myelinoclastic sclerosis, solitary
  • the disorder to be treated is a demyelinating leukostrophic disease including, but not limited to, myelitis, central pontine myelinolysis (CPM), extrapontine myelinolysis, tabes dorsalis, progressive multifocal leukoencephalopathy, leukoencephalopathy with vanishing white matter, leukoencephalopathy with neuroaxonal spheroids, reversible posterior leukoencephalopathy syndrome, megalencephalic leukoencephalopathy with subcortical cysts, megalencephalic leukoencephalopathy with subcortical cysts 1, hypertensive leukoencephalopathy, Metachromatic leukodystrophy, Krabbe disease, Canavan disease, X-linked adrenoleukodystrophy, Alexander disease, cerebrotendineous xanthomatosis, Pelizaeus-Merzbacher disease, Refsum disease.
  • myelitis central pontine myelinolysis (CPM),
  • an active compound as described herein is used to treat Buerger's disease, also known as thromboangiitis obliterans.
  • an active compound as described herein is used to treat giant cell arteritis.
  • an active compound as described herein is used to treat Raynaud's disease.
  • the disorder to be treated is a demyelinating disease of the peripheral nervous system, including, but not limited to, Guillain-Barre syndrome and its chronic counterpart, chronic inflammatory demyelinating polyneuropathy, anti-MAG peripheral neuropathy, Charcot-Marie-Tooth disease and its counterpart Hereditary neuropathy with liability to pressure palsy, Copper deficiency-associated conditions (peripheral neuropathy, myelopathy, and rarely optic neuropathy), and progressive inflammatory neuropathy.
  • demyelinating disease of the peripheral nervous system including, but not limited to, Guillain-Barre syndrome and its chronic counterpart, chronic inflammatory demyelinating polyneuropathy, anti-MAG peripheral neuropathy, Charcot-Marie-Tooth disease and its counterpart Hereditary neuropathy with liability to pressure palsy, Copper deficiency-associated conditions (peripheral neuropathy, myelopathy, and rarely optic neuropathy), and progressive inflammatory neuropathy.
  • the disorder to be treated is a neurological inflammatory disorder.
  • the disorder to be treated is cranial arteritis; giant cell arteritis; Holmes-Adie syndrome; inclusion body myositis (IBM); meningitis; neurologic paraneoplastic syndrome including, but not limited to, Lambert-Eaton myasthenic syndrome, stiff-person syndrome, encephalomyelitis (inflammation of the brain and spinal cord), myasthenia gravis, cerebellar degeneration, limbic and/or brainstem encephalitis, neuromyotonia, and opsoclonus (involving eye movement) and sensory neuropathy; polymyositis; transverse myelitis; vasculitis including temporal arteritis; arachnoiditis; Kinsbourne syndrome or opsoclonus myoclonus syndrome (OMS); or Saint Vitus Dance or sydenham chorea (SD) disease.
  • Lambert-Eaton myasthenic syndrome stiff-person syndrome
  • an active compound or its salt or composition as described herein is used to treat transverse myelitis.
  • the disorder to be treated is a peripheral neuropathy.
  • the peripheral neuropathy is a mononeuropathy.
  • the neuropathy is a polyneuropathy.
  • the polyneuropathy is distal axonopathy, diabetic neuropathy, a demyelinating polyneuropathy, small fiber peripheral neuropathy, mononeuritis multiplex, polyneuritis multiplex, autonomic neuropathy, or neuritis.
  • an active compound or its salt or composition as described herein is used to treat an autoimmune vascular disease.
  • the autoimmune vascular disease is vasculitis.
  • the vasculitis includes, but is not limited to, autoimmune inflammatory vasculitis, Cutaneous small-vessel vasculitis, Granulomatosis with polyangiitis, Eosinophilic granulomatosis with polyangiitis, Behget's disease, Kawasaki disease, Buerger's disease, and “Limited” granulomatosis with polyangiitis vasculitis.
  • an active compound or its salt or composition as described herein is used to treat an arteritis.
  • the arteritis includes, but is not limited to, giant cell arteritis, Takayasu arteritis, temporal arteritis, and polyarteritis nodosa.
  • the complement-mediated disease or disorder comprises transplant rejection. In some embodiments, the complement-mediated disease or disorder is antibody-mediated transplant rejection.
  • an active compound or its salt or composition as described herein is used to treat a proliferative disorder, including, but not limited to, cancer.
  • Targeted cancers suitable for administration of an active compound or its salt described herein include, but are not limited to, estrogen-receptor positive cancer, HER2-negative advanced breast cancer, late-line metastatic breast cancer, liposarcoma, non-small cell lung cancer, liver cancer, ovarian cancer, glioblastoma, refractory solid tumors, retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers, adenocarcinoma of the colon, adenocarcinoma of the rectum, central nervous system germ cell tumors, teratomas, estrogen receptor-negative breast cancer, estrogen receptor-positive breast cancer, familial testicular germ cell tumors, HER2-negative breast cancer, HER2-positive breast cancer, male breast cancer,
  • the targeted cancers included estrogen-receptor positive, HER2-negative advanced breast cancer, late-line metastatic breast cancer, liposarcoma, non-small cell lung cancer, liver cancer, ovarian cancer, glioblastoma, refractory solid tumors, retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers, metastatic colorectal cancer, metastatic melanoma with CDK4 mutation or amplification, or cisplatin-refractory, unresectable germ cell tumors, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix,
  • the methods described herein can be used to treat a host, for example a human, with a lymphoma or lymphocytic or myelocytic proliferation disorder or abnormality.
  • a host for example a human
  • the methods as described herein can be administered to a host with a Hodgkin Lymphoma or a Non-Hodgkin Lymphoma.
  • the host can have a Non-Hodgkin Lymphoma such as, but not limited to: an AIDS-Related Lymphoma; Anaplastic Large-Cell Lymphoma; Angioimmunoblastic Lymphoma; Blastic NK-Cell Lymphoma; Burkitt's Lymphoma; Burkitt-like Lymphoma (Small Non-Cleaved Cell Lymphoma); Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma; Cutaneous T-Cell Lymphoma; Diffuse Large B-Cell Lymphoma; Enteropathy-Type T-Cell Lymphoma; Follicular Lymphoma; Hepatosplenic Gamma-Delta T-Cell Lymphoma; Lymphoblastic Lymphoma; Mantle Cell Lymphoma; Marginal Zone Lymphoma; Nasal T-Cell Lymphoma; Pediatric Lymphoma; Peripheral T-
  • the patient has an acute myelogenous leukemia, for example an undifferentiated AML (MO); myeloblastic leukemia (M1; with/without minimal cell maturation); myeloblastic leukemia (M2; with cell maturation); promyelocytic leukemia (M3 or M3 variant [M3V]); myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]); monocytic leukemia (M5); erythroleukemia (M6); or megakaryoblastic leukemia (M7), small cell lung cancer, retinoblastoma, HPV positive malignancies like cervical cancer and certain head and neck cancers, MYC amplified tumors such as Burkitts' Lymphoma, and triple negative breast cancer; certain classes of sarcoma, certain classes of non-small cell lung carcinoma, certain classes of melanoma, certain classes of pancreatic cancer, certain classes of
  • an active compound or its salt as described herein can be used to preserve or prevent damage to an organ or blood product.
  • an active compound or its salt described herein can be used to prevent damage to an organ, tissue, cell product, or blood product, that has been harvested for transplantation.
  • the organ is the heart, kidney, pancreas, lung, liver, or intestine.
  • the tissue is derived from the cornea, bone, tendon, muscle, heart valve, nerve, artery or vein, or the skin.
  • the blood product is whole blood, plasma, red blood cells or reticulocytes.
  • an active compound or its salt or composition as described herein prevents or delays the onset of at least one symptom of a complement-mediated disease or disorder in an individual.
  • an active compound or its salt or composition as described herein reduces or eliminates at least one symptom of a complement-mediated disease or disorder in an individual.
  • symptoms include, but are not limited to, symptoms associated with autoimmune disease, cancer, hematological disease, infectious disease, inflammatory disease, ischemia-reperfusion injury, neurodegenerative disease, neurodegenerative disorder, renal disease, transplant rejection, ocular disease, vascular disease, or a vasculitis disorder.
  • the symptom can be a neurological symptom, for example, impaired cognitive function, memory impairment, loss of motor function, etc.
  • the symptom can also be the activity of factor D protein in a cell, tissue, or fluid of an individual.
  • the symptom can also be the extent of complement activation in a cell, tissue, or fluid of an individual.
  • administering an active compound or its salt or composition as described herein to an individual modulates complement activation in a cell, tissue, or fluid of an individual. In some embodiments, administration of an active compound or its salt or composition as described herein to an individual inhibits complement activation in a cell, tissue, or fluid of an individual.
  • an active compound or its salt or composition as described herein when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, inhibits complement activation in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to complement activation in the individual before treatment with the compounds described herein.
  • an active compound or its salt or composition as described herein reduces C3 deposition onto red blood cells; for example, in some embodiments, an an active compound or its salt or composition as described herein reduces deposition of C3b, iC3b, etc., onto RBCs. In some embodiments, an active compound or its salt or composition as described herein inhibits complement-mediated red blood cell lysis.
  • an active compound or its salt or composition as described herein reduces C3 deposition onto platelets; for example, in some embodiments, an active compound or its salt or composition as described herein reduces deposition of C3b, iC3b, etc., onto platelets.
  • administering an active compound or its salt or composition as described herein results in an outcome selected from the group consisting of: (a) a reduction in complement activation; (b) an improvement in cognitive function; (c) a reduction in neuron loss; (d) a reduction in phospho-Tau levels in neurons; (e) a reduction in glial cell activation; (f) a reduction in lymphocyte infiltration; (g) a reduction in macrophage infiltration; (h) a reduction in antibody deposition, (i) a reduction in glial cell loss; (j) a reduction in oligodendrocyte loss; (k) a reduction in dendritic cell infiltration; (l) a reduction in neutrophil infiltration; (m) a reduction in red blood cell lysis; (n) a reduction in red blood cell phagocytosis; (o) a reduction in platelet phagocytosis; (p) a reduction in platelet lysis; (q) an improvement in transplant graft survival; (a) a
  • an active compound or its salt or composition as described herein when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, is effect to achieve a reduction of at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, of one or more of the following outcomes: (a) complement activation; (b) decline in cognitive function; (c) neuron loss; (d) phospho-Tau levels in neurons; (e) glial cell activation; (f) lymphocyte infiltration; (g) macrophage infiltration; (h) antibody deposition, (i) glial cell loss; (j) oligodendrocyte loss; (k) dendritic cell infiltration; (1) neutrophil infiltration; (m) red blood cell lysis; (n) red blood cell phagocytosis; (o)
  • an active compound or its salt or composition as described herein when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, is effect to achieve an improvement of at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, of one or more of the following outcomes: a) cognitive function; b) transplant graft survival; c) vision; d) motor control; e) thrombus formation; f) clotting; g) kidney function; and h) hematocrit (red blood cell count), compared to the level or degree of the outcome in the individual before treatment with the active compound.
  • administering an active compound or its salt or composition as described herein to an individual reduces complement activation in the individual.
  • an active compound or its salt or composition as described herein when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces complement activation in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to complement activation in the individual before treatment with the active compound or its salt.
  • administering an active compound or its salt or composition as described herein improves cognitive function in the individual.
  • an active compound described herein when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, improves cognitive function in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the cognitive function in the individual before treatment with the active compound.
  • administering an active compound or its salt or composition as described herein reduces the rate of decline in cognitive function in the individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces the rate of decline of cognitive function in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the rate of decline in cognitive function in the individual before treatment with the active compound or its salt.
  • administering an active compound or its salt or composition as described herein to an individual reduces neuron loss in the individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces neuron loss in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to neuron loss in the individual before treatment with the active compound.
  • administering an active compound or its salt or composition as described herein to an individual reduces phospho-Tau levels in the individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces phospho-Tau in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the phospho-Tau level in the individual before treatment with the active compound or its salt.
  • administering an active compound or its salt or composition as described herein to an individual reduces glial cell activation in the individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces glial activation in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to glial cell activation in the individual before treatment with the active compound or its salt.
  • the glial cells are astrocytes or microglia.
  • administering an active compound or its salt or composition as described herein to an individual reduces lymphocyte infiltration in the individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces lymphocyte infiltration in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to lymphocyte infiltration in the individual before treatment with the active compound or its salt.
  • administering an active compound or its salt or composition as described herein to an individual reduces macrophage infiltration in the individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces macrophage infiltration in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to macrophage infiltration in the individual before treatment with the active compound or its salt.
  • administering an active compound or its salt or composition as described herein to an individual reduces antibody deposition in the individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces antibody deposition in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to antibody deposition in the individual before treatment with the active compound or its salt.
  • administering an active compound or its salt or composition as described herein to an individual reduces anaphylatoxin (e.g., C3a, C4a, C5a) production in an individual.
  • an active compound or its salt when administered in one or more doses as monotherapy or in combination therapy to an individual having a complement-mediated disease or disorder, reduces anaphylatoxin production in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, compared to the level of anaphylatoxin production in the individual before treatment with the active compound or its salt.
  • the present disclosure provides for use of an active compound or its salt of the present disclosure or a pharmaceutical composition comprising an active compound or its salt of the present disclosure and a pharmaceutically acceptable excipient to treat an individual having a complement-mediated disease or disorder.
  • the present disclosure provides for use of an active compound or its salt of the present disclosure to treat an individual having a complement-mediated disease or disorder.
  • the present disclosure provides for use of a pharmaceutical composition comprising an active compound or its salt of the present disclosure and a pharmaceutically acceptable excipient to treat an individual having a complement-mediated disease or disorder.
  • an active compound or its salt or composition as described herein may be provided in combination or alternation with or preceded by, concomitant with or followed by, an effective amount of at least one additional therapeutic agent, for example, for treatment of a disorder listed herein.
  • additional therapeutic agent for example, for treatment of a disorder listed herein.
  • second active agents for such combination therapy are provided below.
  • an active compound or its salt or composition as described herein may be provided in combination or alternation with at least one additional inhibitor of the complement system or a second active compound with a different biological mechanism of action.
  • additional inhibitor of the complement system or a second active compound with a different biological mechanism of action.
  • an active compound or its salt or composition as described herein may be provided together with a protease inhibitor, a soluble complement regulator, a therapeutic antibody (monoclonal or polyclonal), complement component inhibitor, receptor agonist, or siRNA.
  • an active compound described herein is administered in combination or alternation with an antibody against tumor necrosis factor (TNF), including but not limited to infliximab (Remicade), adalimumab, certolizumab, golimumab, or a receptor fusion protein such as etanercept (Embrel).
  • TNF tumor necrosis factor

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
US17/267,362 2018-08-20 2019-08-20 Pharmaceutical compounds for the treatment of complement factor d medical disorders Pending US20230022157A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/267,362 US20230022157A1 (en) 2018-08-20 2019-08-20 Pharmaceutical compounds for the treatment of complement factor d medical disorders

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862765318P 2018-08-20 2018-08-20
US201862727847P 2018-09-06 2018-09-06
US17/267,362 US20230022157A1 (en) 2018-08-20 2019-08-20 Pharmaceutical compounds for the treatment of complement factor d medical disorders
PCT/US2019/047252 WO2020041301A1 (fr) 2018-08-20 2019-08-20 Composés pharmaceutiques pour le traitement de troubles médicaux du facteur d du complément

Publications (1)

Publication Number Publication Date
US20230022157A1 true US20230022157A1 (en) 2023-01-26

Family

ID=69591125

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/267,362 Pending US20230022157A1 (en) 2018-08-20 2019-08-20 Pharmaceutical compounds for the treatment of complement factor d medical disorders

Country Status (4)

Country Link
US (1) US20230022157A1 (fr)
EP (1) EP3841086A4 (fr)
JP (2) JP7538113B2 (fr)
WO (1) WO2020041301A1 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115362162A (zh) * 2020-02-20 2022-11-18 艾其林医药公司 用于治疗补体因子d介导的障碍的杂芳基化合物
WO2022006051A1 (fr) * 2020-06-29 2022-01-06 Anovent Pharmaceutical (U.S.), Llc Formulation de tocilizumab et procédé de traitement de la covid-19 par inhalation
BR112023018321A2 (pt) 2021-03-15 2024-01-02 Maze Therapeutics Inc Inibidores de glicogênio sintase 1 (gys1) e métodos de uso dos mesmos
WO2024008121A1 (fr) * 2022-07-06 2024-01-11 南京明德新药研发有限公司 Composés azabicyclo difluoro-substitués et leurs utilisations
WO2024191606A1 (fr) * 2023-03-13 2024-09-19 Alexion Pharmaceuticals, Inc. Formulations de danicopan et leurs procédés d'utilisation

Family Cites Families (230)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB969808A (en) 1962-06-08 1964-09-16 Boots Pure Drug Co Ltd Anthelmintic compositions and compounds
IT1148784B (it) 1980-04-09 1986-12-03 Eurand Spa Procedimento per la preparazione di microcapsule in un veicolo liquido
US4946929A (en) 1983-03-22 1990-08-07 Massachusetts Institute Of Technology Bioerodible articles useful as implants and prostheses having predictable degradation rates
US4794000A (en) 1987-01-08 1988-12-27 Synthetic Blood Corporation Coacervate-based oral delivery system for medically useful compositions
US4638045A (en) 1985-02-19 1987-01-20 Massachusetts Institute Of Technology Non-peptide polyamino acid bioerodible polymers
US4806621A (en) 1986-01-21 1989-02-21 Massachusetts Institute Of Technology Biocompatible, bioerodible, hydrophobic, implantable polyimino carbonate article
US5736372A (en) 1986-11-20 1998-04-07 Massachusetts Institute Of Technology Biodegradable synthetic polymeric fibrous matrix containing chondrocyte for in vivo production of a cartilaginous structure
US5759830A (en) 1986-11-20 1998-06-02 Massachusetts Institute Of Technology Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
CA1340581C (fr) 1986-11-20 1999-06-08 Joseph P. Vacanti Neomorphogenese chimerique d'organes par implatation cellulaire controlee, utilisant des matrices artificielles
US5019379A (en) 1987-07-31 1991-05-28 Massachusetts Institute Of Technology Unsaturated polyanhydrides
JP2528706B2 (ja) 1988-05-30 1996-08-28 ゼリア新薬工業株式会社 ジヒドロピリジン化合物の製剤組成物
GB8819110D0 (en) 1988-08-11 1988-09-14 Norsk Hydro As Antihypertensive drug & method for production
US5010167A (en) 1989-03-31 1991-04-23 Massachusetts Institute Of Technology Poly(amide-and imide-co-anhydride) for biological application
US5019400A (en) 1989-05-01 1991-05-28 Enzytech, Inc. Very low temperature casting of controlled release microspheres
US5013557A (en) 1989-10-03 1991-05-07 Warner-Lambert Company Taste masking compositions comprising spray dried microcapsules containing sucralfate and methods for preparing same
KR0182801B1 (ko) 1991-04-16 1999-05-01 아만 히데아키 고체 분산체의 제조방법
TW209174B (fr) 1991-04-19 1993-07-11 Takeda Pharm Industry Co Ltd
GB9206757D0 (en) 1992-03-27 1992-05-13 Ferring Bv Novel peptide receptor ligands
AU4198793A (en) 1992-07-24 1994-01-27 Takeda Chemical Industries Ltd. Microparticle preparation and production thereof
US5399665A (en) 1992-11-05 1995-03-21 Massachusetts Institute Of Technology Biodegradable polymers for cell transplantation
US5512600A (en) 1993-01-15 1996-04-30 Massachusetts Institute Of Technology Preparation of bonded fiber structures for cell implantation
US5514378A (en) 1993-02-01 1996-05-07 Massachusetts Institute Of Technology Biocompatible polymer membranes and methods of preparation of three dimensional membrane structures
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5565215A (en) 1993-07-23 1996-10-15 Massachusettes Institute Of Technology Biodegradable injectable particles for imaging
GB9322014D0 (en) 1993-10-26 1993-12-15 Co Ordinated Drug Dev Improvements in and relating to carrier particles for use in dry powder inhalers
SE9401109D0 (sv) 1994-03-31 1994-03-31 Leiras Oy Opthalmic composition II
SE9401108D0 (sv) 1994-03-31 1994-03-31 Leiras Oy Ophthalmic composition I
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
GB9412273D0 (en) 1994-06-18 1994-08-10 Univ Nottingham Administration means
US6007845A (en) 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
US5716404A (en) 1994-12-16 1998-02-10 Massachusetts Institute Of Technology Breast tissue engineering
EP0797987B1 (fr) 1994-12-19 2003-10-15 Daiichi Pharmaceutical Co., Ltd. Preparation granulaire a liberation prolongee et procede de production
GB9501841D0 (en) 1995-01-31 1995-03-22 Co Ordinated Drug Dev Improvements in and relating to carrier particles for use in dry powder inhalers
US6413539B1 (en) 1996-10-31 2002-07-02 Poly-Med, Inc. Hydrogel-forming, self-solvating absorbable polyester copolymers, and methods for use thereof
US6123727A (en) 1995-05-01 2000-09-26 Massachusetts Institute Of Technology Tissue engineered tendons and ligaments
GB9515182D0 (en) 1995-07-24 1995-09-20 Co Ordinated Drug Dev Improvements in and relating to powders for use in dry powder inhalers
ES2199289T3 (es) 1995-07-26 2004-02-16 Kyowa Hakko Kogyo Co., Ltd. Dispersion solida de derivados de xantina.
US6095148A (en) 1995-11-03 2000-08-01 Children's Medical Center Corporation Neuronal stimulation using electrically conducting polymers
US5902599A (en) 1996-02-20 1999-05-11 Massachusetts Institute Of Technology Biodegradable polymer networks for use in orthopedic and dental applications
ATE238068T1 (de) 1996-07-01 2003-05-15 Univ Utrecht Hydrolysierbare hydrogele zur gesteuerten freisetzung
US6395302B1 (en) 1996-11-19 2002-05-28 Octoplus B.V. Method for the preparation of microspheres which contain colloidal systems
EP0842657A1 (fr) 1996-11-19 1998-05-20 OctoPlus B.V. Microsphères pour la libération contrÔlée et procédés pour la préparation de telles microsphères
AU7808198A (en) 1997-06-03 1998-12-21 Biocryst Pharmaceuticals, Inc. Novel compounds useful in the complement, coagulat and kallikrein pathways and method for their preparation
US5837752A (en) 1997-07-17 1998-11-17 Massachusetts Institute Of Technology Semi-interpenetrating polymer networks
ATE364374T1 (de) 1997-08-11 2007-07-15 Pfizer Prod Inc Feste pharmazeutische dispersionen mit erhöhter bioverfügbarkeit
US6686446B2 (en) 1998-03-19 2004-02-03 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6632922B1 (en) 1998-03-19 2003-10-14 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6506577B1 (en) 1998-03-19 2003-01-14 The Regents Of The University Of California Synthesis and crosslinking of catechol containing copolypeptides
US6410561B1 (en) 1998-03-26 2002-06-25 Japan Tobacco Inc. Amide derivatives and nociceptin antagonists
GB9827145D0 (en) 1998-12-09 1999-02-03 Co Ordinated Drug Dev Improvements in or relating to powders
US6287588B1 (en) 1999-04-29 2001-09-11 Macromed, Inc. Agent delivering system comprised of microparticle and biodegradable gel with an improved releasing profile and methods of use thereof
GB9927011D0 (en) 1999-11-16 2000-01-12 Advanced Phytonics Ltd Method for the production of particles
EP1239844B1 (fr) 1999-12-20 2005-06-08 Nicholas J. Kerkhof Procede de production de particules d'echelle nanometrique par sechage par atomisation en lit fluidise
EP1712222A3 (fr) 1999-12-23 2012-06-20 Pfizer Products Inc. Compositions pharmaceutiques fournissant des concentrations de médicaments améliorées
CN1402632A (zh) 1999-12-23 2003-03-12 辉瑞产品公司 水凝胶驱动的药物剂型
EP1267946A4 (fr) 2000-02-28 2008-07-02 Genesegues Inc Systeme et procede d'encapsulation de nanocapsules
CA2404737C (fr) 2000-04-03 2010-06-29 Santen Pharmaceutical Co., Ltd. Substance pour l'administration d'un medicament, contenant du polyalkylene glycol et un phospholipide joints par covalence au medicament
GB0009468D0 (en) 2000-04-17 2000-06-07 Vectura Ltd Improvements in or relating to formulations for use in inhaler devices
GB0009469D0 (en) 2000-04-17 2000-06-07 Vectura Ltd Improvements in or relating to formalities for use in inhaler devices
US6589549B2 (en) 2000-04-27 2003-07-08 Macromed, Incorporated Bioactive agent delivering system comprised of microparticles within a biodegradable to improve release profiles
US6495164B1 (en) 2000-05-25 2002-12-17 Alkermes Controlled Therapeutics, Inc. I Preparation of injectable suspensions having improved injectability
EP1913939B1 (fr) 2000-06-27 2017-05-31 Vectura Limited Formulations destinées à être utilisées dans des dispositifs d'inhalation
GB2364919A (en) 2000-07-21 2002-02-13 Cambridge Consultants Inhalers
GB0021024D0 (en) 2000-08-29 2000-10-11 Glaxo Group Ltd Inhalation device
ATE537845T1 (de) 2000-10-31 2012-01-15 Pr Pharmaceuticals Inc Verfahren zur herstellung von formulierungen zur verbesserten abgabe von bioaktiven molekülen
ES2689704T3 (es) 2000-11-30 2018-11-15 Vectura Limited Partículas para usar en una composición farmacéutica
US6818732B2 (en) 2001-08-30 2004-11-16 The Regents Of The University Of California Transition metal initiators for controlled poly (beta-peptide) synthesis from beta-lactam monomers
GB0122318D0 (en) 2001-09-14 2001-11-07 Novartis Ag Organic compounds
EP1469829B1 (fr) 2002-02-01 2016-01-27 Bend Research, Inc Formes posologiques a liberation immediate, contenant des dispersions medicamenteuses solides
ITMI20021527A1 (it) 2002-07-11 2004-01-12 Consiglio Nazionale Ricerche Anticorpi anti componente c5 del complemento e loro uso
US9415102B2 (en) 2002-09-06 2016-08-16 Alexion Pharmaceuticals, Inc. High concentration formulations of anti-C5 antibodies
EP1567487A4 (fr) 2002-11-15 2005-11-16 Bristol Myers Squibb Co Modulateurs lies a prolyl uree a chaine ouverte de la fonction du recepteur d'androgene
US20050009910A1 (en) 2003-07-10 2005-01-13 Allergan, Inc. Delivery of an active drug to the posterior part of the eye via subconjunctival or periocular delivery of a prodrug
WO2005025550A1 (fr) 2003-09-15 2005-03-24 Vectura Limited Compositions pharmaceutiques permettant de traiter l'ejaculation precoce par inhalation pulmonaire
CA2539324A1 (fr) 2003-09-18 2005-03-31 Macusight, Inc. Administration transsclerale d'agents therapeutiques
ES2383689T3 (es) 2003-09-23 2012-06-25 University Of North Carolina At Chapel Hill Perfluoropoliéteres fotocurables para su uso como materiales novedosos en dispositivos microfluídicos
SE0302665D0 (sv) 2003-10-07 2003-10-07 Astrazeneca Ab Novel Process
WO2005044186A2 (fr) 2003-10-28 2005-05-19 Glaxo Group Limited Formulations pharmaceutiques destinees a etre inhalees utilisant des agents dessechants et methodes d'administration desdites formulations pharmaceutiques
DK1704585T3 (en) 2003-12-19 2017-05-22 Univ North Carolina Chapel Hill Methods for preparing isolated micro- and nanostructures using soft lithography or printing lithography
US8957034B2 (en) 2004-01-28 2015-02-17 Johns Hopkins University Drugs and gene carrier particles that rapidly move through mucous barriers
WO2007021762A2 (fr) 2005-08-09 2007-02-22 The University Of North Carolina At Chapel Hill Procedes et materiaux permettant de fabriquer des dispositifs microfluidiques
GB0407627D0 (en) 2004-04-02 2004-05-05 Vectura Ltd Corkscrew pump
ES2246694B1 (es) 2004-04-29 2007-05-01 Instituto Cientifico Y Tecnologico De Navarra, S.A. Nanoparticulas pegiladas.
US7771742B2 (en) 2004-04-30 2010-08-10 Allergan, Inc. Sustained release intraocular implants containing tyrosine kinase inhibitors and related methods
WO2005107708A1 (fr) 2004-04-30 2005-11-17 Allergan, Inc. Implants d'inhibiteurs de tyrosine kinase intravitreens, biodegradables
US20050244469A1 (en) 2004-04-30 2005-11-03 Allergan, Inc. Extended therapeutic effect ocular implant treatments
PL370285A1 (pl) 2004-09-23 2006-04-03 Glaxosmithkline Pharmaceuticals Spółka Akcyjna Inhalator proszkowy
CA2594694A1 (fr) 2005-01-28 2006-08-03 Pfizer Products Inc. Deshydratation de particules contenant un medicament
EP2548894A1 (fr) 2005-02-03 2013-01-23 Bend Research, Inc. Compositions pharmaceutiques à éfficacité améliorée
US8663639B2 (en) 2005-02-09 2014-03-04 Santen Pharmaceutical Co., Ltd. Formulations for treating ocular diseases and conditions
DK1848431T3 (en) 2005-02-09 2016-04-18 Santen Pharmaceutical Co Ltd LIQUID FORMULATIONS FOR TREATMENT OF DISEASES OR CONDITIONS
AU2006282042B2 (en) 2005-06-17 2011-12-22 The University Of North Carolina At Chapel Hill Nanoparticle fabrication methods, systems, and materials
US20070071756A1 (en) 2005-09-26 2007-03-29 Peyman Gholam A Delivery of an agent to ameliorate inflammation
US8168584B2 (en) 2005-10-08 2012-05-01 Potentia Pharmaceuticals, Inc. Methods of treating age-related macular degeneration by compstatin and analogs thereof
WO2007056561A2 (fr) 2005-11-09 2007-05-18 Liquidia Technologies, Inc. Dispositif medical, materiaux et procedes
WO2007076358A1 (fr) 2005-12-23 2007-07-05 Alcon, Inc. Preparation pharmaceutique pour l'administration, dans l'oeil, de composes inhibiteurs des récepteurs tyrosine kinases (rtki)
WO2007081876A2 (fr) 2006-01-04 2007-07-19 Liquidia Technologies, Inc. Surfaces nanostructurées pour applications biomédicales/biomatérielles et traitements à partir de celles-ci
CA2636716C (fr) 2006-01-13 2014-12-23 Surmodics, Inc. Matrices contenant des microparticules pour l'administration de medicaments
TWI398433B (zh) 2006-02-10 2013-06-11 Dow Agrosciences Llc 殺蟲性之n-取代(6-鹵烷基吡啶-3-基)烷基磺醯亞胺
BRPI0709016A2 (pt) 2006-03-23 2011-06-21 Macusight Inc formulações e métodos para doenças ou condições relacionadas com a permeabilidade vascular
US20080166411A1 (en) 2006-04-10 2008-07-10 Pfizer Inc Injectable Depot Formulations And Methods For Providing Sustained Release Of Poorly Soluble Drugs Comprising Nanoparticles
US20100003291A1 (en) 2006-05-15 2010-01-07 Liquidia Technologies, Inc. Nano-particles for cosmetic applications
WO2008013952A2 (fr) 2006-07-27 2008-01-31 The University Of North Carolina At Chapel Hill Procédés et systèmes de fabrication de nanoparticules, et matériaux permettant de fabriquer des globules rouges artificiels
GB0617480D0 (en) 2006-09-06 2006-10-18 Univ Sheffield Novel nanoparticles
DE602007012559D1 (de) 2006-09-08 2011-03-31 Univ Johns Hopkins H die schleimhaut
US9022970B2 (en) 2006-10-16 2015-05-05 Alcon Research, Ltd. Ophthalmic injection device including dosage control device
EP2409687B1 (fr) 2006-11-09 2014-06-11 Alcon Research, Ltd. Matrice polymère insoluble dans l'eau pour l'administration de médicaments
WO2008100304A2 (fr) 2006-11-15 2008-08-21 The University Of North Carolina At Chapel Hill Composite de particules polymères comportant des particules qui présentent une forme, une taille et un ordre de haute fidélité
PL2148691T3 (pl) 2007-02-05 2015-12-31 Apellis Pharmaceuticals Inc Analogi kompstatyny do stosowania w leczeniu stanów zapalnych układu oddechowego
AU2008314647B2 (en) 2007-10-12 2013-03-21 Massachusetts Institute Of Technology Vaccine nanotechnology
US20090203709A1 (en) 2008-02-07 2009-08-13 Abbott Laboratories Pharmaceutical Dosage Form For Oral Administration Of Tyrosine Kinase Inhibitor
US9943401B2 (en) 2008-04-04 2018-04-17 Eugene de Juan, Jr. Therapeutic device for pain management and vision
WO2009132265A2 (fr) 2008-04-25 2009-10-29 The University Of North Carolina At Chapel Hill Composés dégradables et leurs procédés d'utilisation, impliquant en particulier une répartition des particules dans des matrices non mouillantes
WO2009132206A1 (fr) 2008-04-25 2009-10-29 Liquidia Technologies, Inc. Compositions et procédés pour administration et libération intracellulaire de chargement
WO2010009087A1 (fr) 2008-07-15 2010-01-21 Eyegate Pharmaceuticals, Inc. Administration iontophorétique d'une formulation à libération contrôlée dans l'œil
MX2011001371A (es) 2008-08-05 2011-06-16 Novartis Ag Composiciones y metodos para anticuerpos que se dirigen a la proteina de complementos c5.
UA101056C2 (ru) 2008-10-08 2013-02-25 Астразенека Аб Ингаляционное устройство и способ дозированной подачи медикамента
WO2010065748A2 (fr) 2008-12-05 2010-06-10 Liquidia Technologies, Inc. Procédé de fabrication de matériaux à configuration
US8623395B2 (en) 2010-01-29 2014-01-07 Forsight Vision4, Inc. Implantable therapeutic device
CN104887389B (zh) 2009-01-29 2017-06-23 弗赛特影像4股份有限公司 后段给药
WO2010091187A2 (fr) 2009-02-04 2010-08-12 The Brigham And Women's Hospital, Inc. Nanoparticules polymères dont la charge de médicament est augmentée et procédé d'utilisation
US8192408B2 (en) 2009-02-10 2012-06-05 Psivida Us, Inc. Ocular trocar assembly
JP5653942B2 (ja) 2009-02-26 2015-01-14 ザ ユニバーシティ オブ ノース キャロライナ アット チャペル ヒル 介入薬剤送達システム
GB2469471B (en) 2009-04-14 2015-01-14 Skype Optimising communications
US10952965B2 (en) 2009-05-15 2021-03-23 Baxter International Inc. Compositions and methods for drug delivery
CA2764063C (fr) 2009-06-03 2019-05-14 Forsight Labs, Llc Distribution de medicament de segment anterieur
WO2011008737A2 (fr) 2009-07-13 2011-01-20 The University Of North Carolina At Chapel Hill Particules d'aérosol modifiées et procédés associés
TWI492769B (zh) 2009-09-23 2015-07-21 Alcon Res Ltd 可注射的水性眼用組成物及其使用之方法
NO2490635T3 (fr) 2009-10-23 2018-02-03
EP2490620A4 (fr) 2009-10-23 2017-03-22 Forsight Labs, Llc Écran thérapeutique adaptable pour la vision et la douleur
TWI478730B (zh) 2009-12-03 2015-04-01 Alcon Res Ltd 眼科乳劑
US10166142B2 (en) 2010-01-29 2019-01-01 Forsight Vision4, Inc. Small molecule delivery with implantable therapeutic device
EP2538929A4 (fr) 2010-02-25 2014-07-09 Univ Johns Hopkins Délivrance prolongée d'agents thérapeutiques à un compartiment oculaire
US20130071349A1 (en) 2010-03-02 2013-03-21 Allergan, Inc. Biodegradable polymers for lowering intraocular pressure
AU2011248129B2 (en) 2010-05-05 2014-10-09 Alcon Inc. Stabilized ophthalmic galactomannan formulations
US8999312B2 (en) 2010-06-02 2015-04-07 Alcon Research, Ltd. Use of PBO-PEO-PBO block copolymers in ophthalmic compositions
US20130324482A1 (en) 2010-07-09 2013-12-05 Apellis Pharmaceuticals, Inc. Compstatin analogs for treatment of rhinosinusitis and nasal polyposis
AU2011285548B2 (en) 2010-08-05 2014-02-06 Forsight Vision4, Inc. Combined drug delivery methods and apparatus
SI2600930T1 (sl) 2010-08-05 2021-08-31 Forsight Vision4, Inc. Injekcijska naprava za dajanje zdravila
WO2012019047A2 (fr) 2010-08-05 2012-02-09 Forsight Vision4, Inc. Implant sous-conjonctival pour une administration d'un médicament dans le segment postérieur
HUE057267T2 (hu) 2010-08-05 2022-05-28 Forsight Vision4 Inc Berendezés szem kezelésére
WO2012031129A2 (fr) 2010-09-03 2012-03-08 Bend Research, Inc. Appareil de séchage par pulvérisation et ses procédés d'utilisation
WO2012039979A2 (fr) 2010-09-10 2012-03-29 The Johns Hopkins University Diffusion rapide de grosses nanoparticules polymères dans le cerveau de mammifères
AU2011323250B2 (en) 2010-11-05 2015-11-19 The Johns Hopkins University Compositions and methods relating to reduced mucoadhesion
WO2012065006A2 (fr) 2010-11-11 2012-05-18 Forsight Vision4, Inc. Procédés et appareils de détermination de structures poreuses pour l'administration de médicament
TW201304822A (zh) 2010-11-15 2013-02-01 Vectura Ltd 組成物及用途
WO2012068549A2 (fr) 2010-11-19 2012-05-24 Forsight Vision4, Inc. Formulations d'agents thérapeutiques pour des dispositifs implantés
KR20140027090A (ko) 2011-01-04 2014-03-06 노파르티스 아게 연령-관련 황반 변성 (amd)의 치료에 유용한 인돌 화합물 또는 그의 유사체
US9327037B2 (en) 2011-02-08 2016-05-03 The Johns Hopkins University Mucus penetrating gene carriers
WO2012145801A1 (fr) 2011-04-29 2012-11-01 Jagat Rakesh Kanwar Nanoparticule
US9060938B2 (en) 2011-05-10 2015-06-23 Bend Research, Inc. Pharmaceutical compositions of active agents and cationic dextran polymer derivatives
RU2653439C9 (ru) 2011-05-11 2018-10-16 Апеллис Фармасьютикалс, Инк. Клеточно-реактивные аналоги компстатина, аналоги компстатина длительного действия или аналоги компстатина нацеленного действия и их применение
US8691750B2 (en) 2011-05-17 2014-04-08 Axolabs Gmbh Lipids and compositions for intracellular delivery of biologically active compounds
AU2012272706B2 (en) 2011-06-22 2017-07-06 Apellis Pharmaceuticals, Inc. Methods of treating chronic disorders with complement inhibitors
GB201115874D0 (en) 2011-09-14 2011-10-26 Astrazeneca Ab Inhaler
CN106073986B (zh) 2011-09-14 2019-01-11 弗赛特影像5股份有限公司 治疗患者的眼睛的装置
US9883968B2 (en) 2011-09-16 2018-02-06 Forsight Vision4, Inc. Fluid exchange apparatus and methods
US20150037428A1 (en) 2011-11-29 2015-02-05 The University Of North Carolina At Chapel Hill Geometrically engineered particles and methods for modulating macrophage or immune responses
JP6308679B2 (ja) 2011-12-14 2018-04-11 ザ・ジョンズ・ホプキンス・ユニバーシティー 粘膜浸透が増強されたかまたは炎症が低減されたナノ粒子
SI2797652T1 (sl) 2011-12-27 2019-03-29 Vectura Gmbh Inhalacijska naprava s sistemom za povratno informacijo
WO2013110028A1 (fr) 2012-01-19 2013-07-25 The Johns Hopkins University Formulations de nanoparticules présentant une pénétration améliorée dans les muqueuses
WO2013116061A1 (fr) 2012-02-03 2013-08-08 Forsight Vision4, Inc. Procédés et instrument pour l'insertion et le retrait de dispositifs thérapeutiques
US10857310B2 (en) 2012-03-09 2020-12-08 Vectura Gmbh Mixing channel for an inhalation device and inhalation device
CN104394891B (zh) 2012-03-16 2019-04-16 约翰霍普金斯大学 用于递送活性剂的非线性多嵌段共聚物-药物结合物
CA2867381C (fr) 2012-03-16 2016-09-20 The Johns Hopkins University Formulations a liberation controlee pour l'administration d'inhibiteurs du hif-1
US20140107025A1 (en) 2012-04-16 2014-04-17 Jade Therapeutics, Llc Ocular drug delivery system
CA2871745C (fr) 2012-05-03 2023-01-24 Kala Pharmaceuticals, Inc. Nanoparticules pharmaceutiques presentant un transport muqueux ameliore
US9827191B2 (en) 2012-05-03 2017-11-28 The Johns Hopkins University Compositions and methods for ophthalmic and/or other applications
JP6360039B2 (ja) 2012-05-03 2018-07-18 カラ ファーマシューティカルズ インコーポレイテッド 複数の被覆された粒子を含む組成物、医薬組成物、医薬製剤、及び当該粒子の形成方法
CA2872519C (fr) 2012-05-04 2017-09-05 The Johns Hopkins University Vehicules de medicament a base de lipide pour penetration rapide a travers les revetements de mucus
WO2013164802A1 (fr) 2012-05-04 2013-11-07 Novartis Ag Modulateurs des voies du complément et leurs utilisations
EP2864322B1 (fr) 2012-06-20 2016-04-27 Novartis AG Modulateur de la voie d'activation du complément et ses utilisations
EP2867224B1 (fr) 2012-06-28 2017-07-26 Novartis AG Dérivés pyrrolidine et leur utilisation en tant que modulateurs de la voie du complément
WO2014002054A1 (fr) 2012-06-28 2014-01-03 Novartis Ag Dérivés pyrrolidine et leur utilisation en tant que modulateurs de la voie du complément
US9487483B2 (en) 2012-06-28 2016-11-08 Novartis Ag Complement pathway modulators and uses thereof
WO2014002052A1 (fr) 2012-06-28 2014-01-03 Novartis Ag Dérivés pyrrolidine et leur utilisation en tant que modulateurs de la voie du complément
CA2876993A1 (fr) 2012-06-28 2014-01-03 Novartis Ag Derives de pyrrolidine et leur utilisation en tant que modulateurs des voies du complement
US9815819B2 (en) 2012-06-28 2017-11-14 Novartis Ag Complement pathway modulators and uses thereof
WO2014005150A1 (fr) 2012-06-29 2014-01-03 Novartis Ag Formes cristallines du l-(2-((1r,3s,5r)-3-((2-fluoro-3-(trifluorométhoxy)phényl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoéthyl)-5-méthyl-1h-pyrazolo[3,4-c]pyridine-3-carboxamide et sels de celui-ci
WO2014002059A1 (fr) 2012-06-29 2014-01-03 Novartis Ag Formes cristallines de 1-(2-((1r,3s,5r)-3-(((r)-1-(3-chloro-2- fluorophényl)éthyl)carbamoyl)-2-azabicyclo[3.1.0]hexan-2-yl)-2-oxoéthyl)-1h-pyrazolo[3,4-c]pyridine-3-carboxamide
US9550755B2 (en) 2012-07-12 2017-01-24 Novartis Ag Complement pathway modulators and uses thereof
ES1077714Y (es) 2012-07-31 2012-12-20 Gutierrez Jose Ramon Perez Envase de madera
EA201590586A1 (ru) 2012-09-17 2015-08-31 Бинд Терапьютикс, Инк. Терапевтические наночастицы, включающие терапевтический агент, способы их получения и применения
AU2013317899A1 (en) 2012-09-20 2015-05-07 Akina, Inc. Biodegradable microcapsules containing filling material
WO2014066775A1 (fr) 2012-10-26 2014-05-01 Forsight Vision5, Inc. Système ophtalmique pour la libération prolongée de médicament dans l'œil
SI2724741T1 (sl) 2012-10-26 2017-10-30 Vectura Gmbh Inhalacijska naprava za uporabo v terapiji z aerosolom
EP3660033B9 (fr) 2012-11-15 2022-06-22 Apellis Pharmaceuticals, Inc. Analogues de la compstatine à longue durée d'action et compositions et méthodes associées
US20160194359A1 (en) 2012-11-15 2016-07-07 Apellis Pharmaceuticals, Inc. Cell-reactive, long-acting, or targeted compstatin analogs and related compositions and methods
WO2014152959A1 (fr) 2013-03-14 2014-09-25 Forsight Vision4, Inc. Systèmes pour l'administration intra-oculaire entretenue de composés à faible solubilité provenant d'un implant de système de pose d'orifice
CN105229003B (zh) 2013-03-14 2017-03-15 诺华股份有限公司 作为补体因子b抑制剂用于治疗眼科疾病的2‑(1h‑吲哚‑4‑基甲基)‑3h‑咪唑并[4,5‑b]吡啶‑6‑甲腈衍生物
WO2014152391A1 (fr) 2013-03-15 2014-09-25 Apellis Pharmaceuticals, Inc. Analogues de compstatine pénétrant dans les cellules et leurs utilisations
EP4302736A3 (fr) 2013-03-28 2024-04-03 ForSight Vision4, Inc. Implant ophtalmique pour l'administration de substances thérapeutiques
US9504653B2 (en) 2013-04-01 2016-11-29 Allergan, Inc. Microsphere drug delivery system for sustained intraocular release
JO3425B1 (ar) 2013-07-15 2019-10-20 Novartis Ag مشتقات البابيريدينيل-اندول واستخدامها كعامل متمم لمثبطات b
MX2016000675A (es) 2013-07-18 2016-05-10 Novartis Ag Derivados de amino-metil-biarilo como inhibidores del factor de complemento d y usos de los mismos.
WO2015057554A1 (fr) 2013-10-15 2015-04-23 Forsight Vision5, Inc. Préparations et méthodes permettant d'augmenter ou de réduire le mucus
US9676728B2 (en) 2013-10-30 2017-06-13 Novartis Ag 2-benzyl-benzimidazole complement factor B inhibitors and uses thereof
WO2015085234A1 (fr) 2013-12-06 2015-06-11 Forsight Vision4, Inc. Dispositifs thérapeutiques implantables
MX2016007345A (es) 2013-12-06 2016-12-09 Envisia Therapeutics Inc Implante intracameral para el tratamiento de una condicion ocular.
JP2017508788A (ja) 2014-02-25 2017-03-30 アキリオン ファーマシューティカルズ,インコーポレーテッド 補体媒介障害の治療のためのエーテル化合物
MX2016011468A (es) 2014-03-07 2017-01-23 Biocryst Pharm Inc Inhibidores de calicreína plasmática humana.
NZ711451A (en) 2014-03-07 2016-05-27 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
WO2016088082A1 (fr) 2014-12-05 2016-06-09 Novartis Ag Dérivés d'aminométhyl-biaryle en tant qu'inhibiteurs du facteur d du complément et leurs utilisations
EP3331906A1 (fr) 2015-08-06 2018-06-13 Dana-Farber Cancer Institute, Inc. Dégradation modulable de protéine endogène
AR106018A1 (es) 2015-08-26 2017-12-06 Achillion Pharmaceuticals Inc Compuestos de arilo, heteroarilo y heterocíclicos para el tratamiento de trastornos médicos
WO2017035413A2 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés carbamate, ester, et cétone pour le traitement de troubles immunitaires et inflammatoires
WO2017035409A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés aryle, hétéroaryle, et hétérocycliques pour le traitement de troubles immunitaires et inflammatoires
WO2017035417A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés de phosphonate pour le traitement de troubles immunitaires et inflammatoires
WO2017035352A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés carbamate, ester, et cétone pour le traitement de troubles médicaux
WO2017035351A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés amino pour le traitement de troubles médicaux
WO2017035362A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Utilisation de composés inhibiteurs de la voie du complément pour atténuer des réponses immunitaires indésirables associées à une thérapie adoptive par lymphocytes t
WO2017035361A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés disubstitués destinés au traitement de troubles médicaux
AR105808A1 (es) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc Compuestos de amida para el tratamiento de trastornos médicos
WO2017035401A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés amide pour le traitement de troubles immunitaires et inflammatoires
WO2017035415A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés alcyne pour le traitement de troubles immunitaires et inflammatoires
ES2908479T3 (es) 2015-08-26 2022-04-29 Achillion Pharmaceuticals Inc Compuestos para el tratamiento de trastornos inmunitarios e inflamatorios
WO2017035355A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés éther pour le traitement de troubles médicaux
AR105809A1 (es) 2015-08-26 2017-11-08 Achillion Pharmaceuticals Inc Compuestos para el tratamiento de trastornos médicos
WO2017035411A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés d'éther pour le traitement de troubles immunitaires et inflammatoires
WO2017035405A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés amino pour le traitement de troubles immunitaires et inflammatoires
WO2017035348A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés alcyne pour le traitement de troubles médicaux
WO2017035357A1 (fr) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Composés phosphonate destinés au traitement de troubles médicaux
MY198346A (en) 2015-10-01 2023-08-28 Biocryst Pharm Inc Human plasma kallikrein inhibitors
EP3386504A4 (fr) * 2015-12-11 2019-05-22 Lifesci Pharmaceuticals, Inc. Composés inhibiteurs thérapeutiques
TWI747873B (zh) 2016-02-01 2021-12-01 美商百歐克斯製藥公司 苯并吡唑化合物及其類似物
WO2018015818A2 (fr) 2016-07-15 2018-01-25 Lifesci Pharmaceuticals, Inc. Composés inhibiteurs thérapeutiques
IL294069B2 (en) 2017-03-01 2023-11-01 Achillion Pharmaceuticals Inc Aryl, heteroaryl and heterocyclic pharmaceutical compounds for the treatment of medical disorders
WO2018160892A1 (fr) 2017-03-01 2018-09-07 Achillion Pharmaceuticals, Inc. Composés macrocycliques destinés au traitement de troubles médicaux
WO2018160891A1 (fr) 2017-03-01 2018-09-07 Achillion Pharmaceutical, Inc. Composés pharmaceutiques destinés au traitement de troubles médicaux
CN111163767A (zh) 2017-08-02 2020-05-15 艾其林医药公司 治疗阵发性睡眠性血红蛋白尿症的治疗方案

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Wermuth, Molecular variations based in isosteric replacements, The Practice of Medicinal Chemistry, 1996, 203-237 (Year: 1996) *

Also Published As

Publication number Publication date
EP3841086A4 (fr) 2022-07-27
EP3841086A1 (fr) 2021-06-30
JP2024069504A (ja) 2024-05-21
WO2020041301A1 (fr) 2020-02-27
JP7538113B2 (ja) 2024-08-21
JP2021535112A (ja) 2021-12-16

Similar Documents

Publication Publication Date Title
US11926617B2 (en) Aryl, heteroaryl, and heterocyclic compounds for treatment of immune and inflammatory disorders
US11649223B2 (en) Amino compounds for treatment of immune and inflammatory disorders
US11649229B2 (en) Amide compounds for treatment of immune and inflammatory disorders
AU2018227849B2 (en) Aryl, heteroaryl, and heterocyclic pharmaceutical compounds for treatment of medical disorders
US10138225B2 (en) Amide compounds for treatment of medical disorders
US20180201580A1 (en) Compounds for treatment of immune inflammatory disorders
EP3589287B1 (fr) Composés macrocycliques destinés au traitement de troubles médicaux
WO2018160891A1 (fr) Composés pharmaceutiques destinés au traitement de troubles médicaux
WO2017035417A1 (fr) Composés de phosphonate pour le traitement de troubles immunitaires et inflammatoires
WO2017035413A2 (fr) Composés carbamate, ester, et cétone pour le traitement de troubles immunitaires et inflammatoires
JP7538113B2 (ja) 補体d因子の医学的障害の治療のための医薬化合物
US20230085372A1 (en) Pharmaceutical compounds for the treatment of complement mediated disorders
US11814391B2 (en) Macrocyclic compounds for the treatment of medical disorders

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: ACHILLION PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WILES, JASON ALLAN;GADHACHANDA, VENKAT RAO;EASTMAN, KYLE J.;AND OTHERS;SIGNING DATES FROM 20190923 TO 20191003;REEL/FRAME:058936/0353

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED